EP2968549B1 - Methods of cell culture - Google Patents

Methods of cell culture Download PDF

Info

Publication number
EP2968549B1
EP2968549B1 EP14769180.2A EP14769180A EP2968549B1 EP 2968549 B1 EP2968549 B1 EP 2968549B1 EP 14769180 A EP14769180 A EP 14769180A EP 2968549 B1 EP2968549 B1 EP 2968549B1
Authority
EP
European Patent Office
Prior art keywords
glycans
dmso
level
cell
preparation
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP14769180.2A
Other languages
German (de)
French (fr)
Other versions
EP2968549A4 (en
EP2968549B2 (en
EP2968549A1 (en
Inventor
Holly Prentice
Rasheed Tijani
Brett Belongia
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Momenta Pharmaceuticals Inc
Original Assignee
Momenta Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=51528764&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP2968549(B1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Momenta Pharmaceuticals Inc filed Critical Momenta Pharmaceuticals Inc
Publication of EP2968549A1 publication Critical patent/EP2968549A1/en
Publication of EP2968549A4 publication Critical patent/EP2968549A4/en
Application granted granted Critical
Publication of EP2968549B1 publication Critical patent/EP2968549B1/en
Publication of EP2968549B2 publication Critical patent/EP2968549B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/005Glycopeptides, glycoproteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0681Cells of the genital tract; Non-germinal cells from gonads
    • C12N5/0682Cells of the female genital tract, e.g. endometrium; Non-germinal cells from ovaries, e.g. ovarian follicle cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/34Sugars
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/60Buffer, e.g. pH regulation, osmotic pressure
    • C12N2500/62DMSO
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production

Definitions

  • the invention relates generally to cell culture methods.
  • WO2011/061275A1 describes a method of producing glycoproteins in a medium comprising DMSO.
  • Therapeutic polypeptides are an important class of therapeutic biotechnology products, and therapeutic antibodies (including murine, chimeric, humanized and human antibodies and fragments thereof) account for the majority of therapeutic biologic products.
  • the invention provides a method of producing a recombinant protein preparation, the method comprising:
  • the disclosure features a method of producing a recombinant protein preparation having a target value of one or more of high mannose glycans and fucosylated glycans, the method comprising: (a) providing a cell genetically engineered to express a recombinant protein; (b) culturing the cell in a culture medium comprising (e.g., supplemented with) DMSO under conditions in which the cell expresses the recombinant protein; and (c) harvesting (e.g., purifying or isolating from the cell and/or culture medium) a preparation of the recombinant protein produced by the cell, wherein the preparation has the target value of the one or more of high mannose glycans and fucosylated glycans.
  • a culture medium comprising (e.g., supplemented with) DMSO under conditions in which the cell expresses the recombinant protein
  • harvesting e.g., purifying or isolating from the
  • the culture medium comprises DMSO for a time and in an amount effective to modify (e.g., increase or decrease) one or more of the high mannose glycans and fucosylated glycans of the recombinant protein.
  • the culture medium comprises about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO.
  • 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2%
  • the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO.
  • the target value is a level of one or more of high mannose glycans and fucosylated glycans in a reference therapeutic product. In some instances, the target value is a level of one or more of high mannose glycans and fucosylated glycans in a reference therapeutic antibody product. In some instances, the target value is a predetermined pharmaceutical product specification or a quality control criterion for a pharmaceutical preparation, e.g., a Certificate of Analysis (CofA), a Certificate of Testing (CofT), or a Master Batch Record. In some instances, the product specification is a product description in an FDA label, a Physician's Insert, a USP monograph, or an EP monograph.
  • the reference therapeutic product is selected from the group consisting of: abatacept, abciximab, adalimumab, aflibercept, alefacept, alemtuzumab, basiliximab, bevacizumab, belatacept, certolizumab, cetuximab, daclizumab, eculizumab, efalizumab, entanercept, gemtuzumab, ibritumomab, infliximab, muromonab-CD3, natalizumab, omalizumab, palivizumab; panitumumab, ranibizumab, rilonacept, rituximab, tositumomab, and trastuzumab.
  • the target value is one or more of: (a) at least about 0.1% to about 20% high mannose glycans, e.g., at least about 0.1%, about 0.5%, about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, high mannose glycans; and (b) at least about 70% to 100% fucosylated glycans, e.g., at least about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% fucosylated glycans.
  • High mannose glycans can be, e.g., HM3, HM4, HM5, HM6, HM7, HM8, HM9, or combinations thereof.
  • the target value of the one or more of high mannose glycans and fucosylated glycans is higher than a corresponding level in a preparation produced by culturing the cell in the medium not comprising DMSO. In some instances, the target value is higher than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level.
  • the method further comprises evaluating the level of one or more of high mannose glycans and fucosylated glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • MSDS Material Safety Data Sheet
  • CofT Certificate of Testing
  • CofA Certificate of Analysis
  • the disclosure features a method of producing a recombinant protein preparation, the method comprising: (a) providing a target value of one or more of high mannose glycans and fucosylated glycans; (b) providing a cell genetically engineered to express a recombinant protein; (c) culturing the cell in a culture medium comprising (e.g., supplemented with) DMSO under conditions in which the cell expresses the recombinant protein; (d) harvesting a preparation of the recombinant protein produced by the cell; and (e) processing (e.g., one or more of formulating, filling into a container, labeling, packaging) the preparation into a drug product if the preparation meets the target value of the one or more of high mannose glycans and fucosylated glycans.
  • processing e.g., one or more of formulating, filling into a container, labeling, packaging
  • the culture medium comprises DMSO for a time and in an amount effective to modify (e.g., increase or decrease) one or more of the high mannose glycans and fucosylated glycans of the recombinant protein.
  • the method further comprises evaluating the level of one or more of high mannose glycans and fucosylated glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, or Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • MSDS Material Safety Data Sheet
  • CofT Certificate of Testing
  • CofA Certificate of Analysis
  • the culture medium comprises about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO.
  • 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2%
  • the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO..
  • the target value is a level of one or more of high mannose glycans and fucosylated glycans in a reference therapeutic product. In some instances, the target value is a level of one or more of high mannose glycans and fucosylated glycans in a reference therapeutic antibody product. In some instances, the target value is a predetermined pharmaceutical product specification or a quality control criterion for a pharmaceutical preparation, e.g., a Certificate of Analysis (CofA), a Certificate of Testing (CofT), or a Master Batch Record. In some instances, the product specification is a product description in an FDA label, a Physician's Insert, a USP monograph, or an EP monograph.
  • the reference therapeutic product is selected from the group consisting of: abatacept, abciximab, adalimumab, aflibercept, alefacept, alemtuzumab, basiliximab, bevacizumab, belatacept, certolizumab, cetuximab, daclizumab, eculizumab, efalizumab, entanercept, gemtuzumab, ibritumomab, infliximab, muromonab-CD3, natalizumab, omalizumab, palivizumab; panitumumab, ranibizumab, rilonacept, rituximab, tositumomab, and trastuzumab.
  • the target value is one or more of: (a) at least about 0.1% to about 20% high mannose glycans, e.g., at least about 0.1%, about 0.5%, about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, high mannose glycans; and (b) at least about 70% to 100% fucosylated glycans, e.g., at least about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% fucosylated glycans.
  • High mannose glycans can be, e.g., HM3, HM4, HM5, HM6, HM7, HM8, HM9, or combinations thereof.
  • the target value of the one or more of high mannose glycans and fucosylated glycans is higher than a corresponding level in a preparation produced by culturing the cell in the medium not comprising DMSO. In some instances, the target value is higher than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level.
  • the disclosure features a method of increasing a level of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans in a recombinant protein preparation, the method comprising: (a) providing a cell genetically engineered to express a recombinant protein; (b) culturing the cell in a culture medium comprising (e.g., supplemented with) DMSO under conditions in which the cell expresses the recombinant protein; and (c) harvesting a preparation of the recombinant protein produced by the cell, wherein the preparation has an increased level of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans relative to a corresponding level in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising DMSO.
  • the culture medium comprises DMSO for a time and in an amount effective to increase one or more of the high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans of the recombinant protein.
  • the method further comprises processing, (e.g., one or more of formulating, filling into a container, labeling, packaging) the preparation into a drug product if the preparation meets a target value of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans.
  • the culture medium comprises about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO.
  • 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2%
  • the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO..
  • the method further comprises measuring a level of the one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • MSDS Material Safety Data Sheet
  • CofT Certificate of Testing
  • CofA Certificate of Analysis
  • the increased level of the one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans is higher than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level.
  • the increased level is one or more of: (a) at least about 0.1% to about 20% high mannose glycans, e.g., at least about 0.1%, about 0.5%, about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, or more, high mannose glycans; (b) at least about 70% to 100% fucosylated glycans, e.g., at least about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% fucosylated glycans; and (c) at least about 10% to about 90% sialylated glycans, e.g., at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% sialylated glycans
  • the disclosure features a method of decreasing a level of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans in a recombinant protein preparation, the method comprising: (a) providing a cell genetically engineered to express a recombinant protein; (b) culturing the cell in a culture medium comprising a reduced level of DMSO under conditions in which the cell expresses the recombinant protein; and (c) harvesting a preparation of the recombinant protein produced by the cell, wherein the preparation has a decreased level of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans relative to a corresponding level in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising the reduced level of DMSO.
  • the culture medium comprises the reduced level of DMSO for a time and in an amount effective to decrease a level of one or more of the high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans of the recombinant protein.
  • the method further comprises processing, (e.g., one or more of formulating, filling into a container, labeling, packaging) the preparation into a drug product if the preparation meets a target value of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans.
  • the medium comprises about 0.5 to 5% DMSO such as about 5%, 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3%, 2.9%, 2.8%, 2.7%, 2.6%, 2.5%, 2.4%, 2.3%, 2.2%, 2.1%, 2%, 1.9%, 1.8%, 1.7%, 1.6%, 1.5%, 1.4%, 1.3%, 1.2%, 1.1%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5% DMSO.
  • the method further comprises measuring a level of the one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • MSDS Material Safety Data Sheet
  • CofT Certificate of Testing
  • CofA Certificate of Analysis
  • the decreased level of the one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans is lower than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%, of the corresponding level.
  • the decreased level is one or more of: (a) less than about 20% high mannose glycans; (b) less than about 90% fucosylated glycans; and (c) less than about 90% sialylated glycans.
  • High mannose glycans can be, e.g., HM3, HM4, HM5, HM6, HM7, HM8, HM9, or combinations thereof.
  • the disclosure features a method of decreasing a level of one or more fucosylated glycans in a recombinant protein preparation, the method comprising: (a) providing a cell genetically engineered to express a recombinant protein; (b) culturing the cell in a culture medium comprising (e.g., supplemented with) DMSO under conditions in which the cell expresses the recombinant protein; and (c) harvesting a preparation of the recombinant protein produced by the cell, wherein the preparation has a decreased level of one or more fucosylated glycans relative to a corresponding level in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising DMSO.
  • the culture medium comprises DMSO for a time and in an amount effective to decrease the level of one or more fucosylated glycans of the recombinant protein.
  • the method further comprises processing, (e.g., one or more of formulating, filling into a container, labeling, packaging) the preparation into a drug product if the preparation meets a target value of one or more fucosylated glycans.
  • the culture medium comprises about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO.
  • 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2%
  • the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO..
  • the method further comprises measuring a level of fucosylated glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • MSDS Material Safety Data Sheet
  • CofT Certificate of Testing
  • CofA Certificate of Analysis
  • the decreased level of the fucosylated glycans is lower than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%, of the corresponding level. In some instances, the decreased level is less than about 100% fucosylated glycans. In some instances, the one or more fucosylated glycans comprise G0F glycans.
  • the disclosure features a method of increasing a level of one or more fucosylated glycans in a recombinant protein preparation, the method comprising: (a) providing a cell genetically engineered to express a recombinant protein; (b) culturing the cell in a culture medium comprising a reduced level of DMSO under conditions in which the cell expresses the recombinant protein; and (c) harvesting a preparation of the recombinant protein produced by the cell, wherein the preparation has an increased level of one or more fucosylated glycans relative to a corresponding level in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising the reduced level of DMSO.
  • the culture medium comprises a reduced level of DMSO for a time and in an amount effective to increase the level of one or more fucosylated glycans of the recombinant protein.
  • the method further comprises processing, (e.g., one or more of formulating, filling into a container, labeling, packaging) the preparation into a drug product if the preparation meets a target value of one or more fucosylated glycans.
  • the medium comprises about 0.5 to about 5% DMSO
  • the method further comprises measuring a level of fucosylated glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • MSDS Material Safety Data Sheet
  • CofT Certificate of Testing
  • CofA Certificate of Analysis
  • the increased level of the fucosylated glycans is higher than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level.
  • the increased level is at least about 70% to 100% fucosylated glycans, e.g., at least about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% fucosylated glycans.
  • the one or more fucosylated glycans comprise G0F glycans.
  • the culturing step comprises a first stage and a second stage.
  • the first stage comprises culturing the cell in a first culture medium comprising a first level of DMSO
  • the second stage comprises culturing the cell in a second culture medium comprising a second level of DMSO.
  • the first culture medium comprises a reduced level of DMSO relative to the second culture medium
  • the second culture medium comprises an elevated level of DMSO relative to the first culture medium.
  • the first culture medium comprises an elevated level of DMSO (e.g., about 1% to about 5% DMSO)
  • the second culture medium comprises a reduced level of (e.g., does not comprise) DMSO.
  • the first stage comprises culturing the cell in the first culture medium for about 1 to about 8 days, e.g., 1-7, 1-6, 1-5 days.
  • the second stage comprises culturing the cell in the second culture medium for about 1 to about 12 days, e.g., 1-10, 1-9, 1-8, 1-7, 1-6 days.
  • the first stage is a growth stage.
  • the second stage is a production stage.
  • the culture medium further comprises one or more of lysine, cysteine, ammonium, manganese, copper, cobalt, putrescine, a peptone, glucose, galactose, glucosamine, glutamine, a lipid (e.g., cholesterol), and dextran sulfate.
  • the cell is a mammalian cell.
  • the mammalian cell is a CHO (e.g., CHO-K1, DG44, CHO-DXB11, CHOK1SV, CHO-S) Vero, BHK, HeLa, COS, MDCK, or HEK-293 cell.
  • CHO e.g., CHO-K1, DG44, CHO-DXB11, CHOK1SV, CHO-S
  • Vero BHK, HeLa, COS, MDCK, or HEK-293 cell.
  • the recombinant protein is a recombinant therapeutic product. In some instances, the recombinant protein is a recombinant therapeutic antibody product. In some instances, the recombinant protein is a recombinant therapeutic fusion protein.
  • the recombinant protein is abatacept, abciximab, adalimumab, aflibercept, alefacept, alemtuzumab, basiliximab, bevacizumab, belatacept, certolizumab, cetuximab, daclizumab, eculizumab, efalizumab, entanercept, gemtuzumab, ibritumomab, infliximab, muromonab-CD3, natalizumab, omalizumab, palivizumab; panitumumab, ranibizumab, rilonacept, rituximab, tositumomab, and trastuzumab.
  • the disclosure features a preparation of a recombinant protein produced using a method described herein.
  • the disclosure features a method of producing a recombinant therapeutic antibody preparation (e.g., abciximab, adalimumab, alemtuzumab, basiliximab, bevacizumab, certolizumab, cetuximab, daclizumab, eculizumab, efalizumab, gemtuzumab, ibritumomab, infliximab, muromonab-CD3, natalizumab, omalizumab, palivizumab, panitumumab, ranibizumab, rituximab, tositumomab, or trastuzumab), the method comprising: (a) providing a target value (e.g., a predetermined pharmaceutical product specification or a quality control criterion for a pharmaceutical preparation, e.g., a Certificate of Analysis (CofA), a Certificate of Testing (CofA),
  • preparations of polypeptides e.g., antibodies
  • glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • DMSO dimethylsulfoxide
  • culturing the cells in medium comprising DMSO does not affect cell growth, cell viability, or titer.
  • the present disclosure encompasses preparations of polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans), methods of making such polypeptides (e.g., antibodies), and methods of using such polypeptides (e.g., antibodies).
  • polypeptides e.g., antibodies
  • targeted levels of glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • methods of making such polypeptides e.g., antibodies
  • methods of using such polypeptides e.g., antibodies.
  • purified refers to a nucleic acid sequence (e.g., a polynucleotide) or an amino acid sequence (e.g., a polypeptide) that is substantially free of other components.
  • a purified polynucleotide or purified polypeptide is removed or separated from other components present in its natural environment.
  • an isolated polypeptide is one that is separated from other components of a cell in which it was produced (e.g., the endoplasmic reticulum or cytoplasmic proteins and RNA).
  • An isolated polynucleotide is one that is separated from other nuclear components (e.g., histones) and/or from upstream or downstream nucleic acid sequences.
  • An isolated nucleic acid sequence or amino acid sequence can be at least 60% free, or at least 75% free, or at least 90% free, or at least 95% free from other components present in natural environment of the indicated nucleic acid sequence or amino acid sequence.
  • polynucleotide refers to an oligonucleotide, nucleotide, or polynucleotide, and fragments or portions thereof, and to DNA and RNA of genomic or synthetic origin, which may be single- or double- stranded, and represent the sense or anti-sense strand.
  • polypeptide refers to an oligopeptide, peptide, polypeptide, or protein sequence, and fragments or portions thereof, and to naturally occurring or synthetic molecules.
  • amino acid sequence and like terms, such as “polypeptide” or “protein”, are not meant to limit the indicated amino acid sequence to the complete, native amino acid sequence associated with the recited protein molecule.
  • pharmaceutically effective amount refers to an amount (e.g., dose) effective in treating a patient, having a disorder or condition described herein. It is also to be understood herein that a “pharmaceutically effective amount” may be interpreted as an amount giving a desired therapeutic effect, either taken in one dose or in any dosage or route, taken alone or in combination with other therapeutic agents.
  • treatment refers to administering a therapy in an amount, manner, and/or mode effective to improve a condition, symptom, or parameter associated with a disorder or condition or to prevent or reduce progression of a disorder or condition, to a degree detectable to one skilled in the art.
  • An effective amount, manner, or mode can vary depending on the subject and may be tailored to the subject.
  • an antibody refers to a polypeptide that includes at least one immunoglobulin variable region, e.g., an amino acid sequence that provides an immunoglobulin variable domain or immunoglobulin variable domain sequence.
  • an antibody can include a heavy (H) chain variable region (abbreviated herein as VH), and a light (L) chain variable region (abbreviated herein as VL).
  • VH heavy chain variable region
  • L light chain variable region
  • an antibody includes two heavy (H) chain variable regions and two light (L) chain variable regions.
  • antibody encompasses antigen-binding fragments of antibodies (e.g., single chain antibodies, Fab, F(ab') 2 , Fd, Fv, and dAb fragments) as well as complete antibodies, e.g., intact immunoglobulins of types IgA, IgG, IgE, IgD, IgM (as well as subtypes thereof).
  • the light chains of the immunoglobulin can be of types kappa or lambda.
  • an antibody includes an Fc region.
  • an antibody is a therapeutic antibody.
  • Fc region refers to a dimer of two "Fc polypeptides", each "Fc polypeptide” comprising the constant region of an antibody excluding the first constant region immunoglobulin domain.
  • an "Fc region” includes two Fc polypeptides linked by one or more disulfide bonds, chemical linkers, or peptide linkers.
  • Fc polypeptide refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, and the last three constant region immunoglobulin domains of IgE and IgM, and may also include part or all of the flexible hinge N-terminal to these domains.
  • Fc polypeptide comprises immunoglobulin domains Cgamma2 (C ⁇ 2) and Cgamma3 (C ⁇ 3) and the lower part of the hinge between Cgamma1 (C ⁇ 1) and C ⁇ 2.
  • the human IgG heavy chain Fc polypeptide is usually defined to comprise residues starting at T223 or C226 or P230, to its carboxyl-terminus, wherein the numbering is according to the EU index as in Kabat et al. (1991, NIH Publication 91-3242, National Technical Information Services, Springfield, VA).
  • Fc polypeptide comprises immunoglobulin domains Calpha2 (C ⁇ 2) and Calpha3 (C ⁇ 3) and the lower part of the hinge between Calpha1 (C ⁇ 1) and C ⁇ 2.
  • An Fc region can be synthetic, recombinant, or generated from natural sources such as IVIG.
  • a "glycan” is a sugar.
  • Glycans can be monomers or polymers of sugar residues, but typically contain at least three sugars, and can be linear or branched.
  • a glycan may include natural sugar residues (e.g., glucose, N-acetylglucosamine, N-acetyl neuraminic acid, galactose, mannose, fucose, hexose, arabinose, ribose, xylose, etc.) and/or modified sugars (e.g., 2'-fluororibose, 2'-deoxyribose, phosphomannose, 6'-sulfo N-acetylglucosamine, etc).
  • natural sugar residues e.g., glucose, N-acetylglucosamine, N-acetyl neuraminic acid, galactose, mannose, fucose, hexose, arabinose, ribo
  • glycocan includes homo and heteropolymers of sugar residues.
  • glycan also encompasses a glycan component of a glycoconjugate (e.g., of a glycoprotein, glycolipid, proteoglycan, etc.).
  • a glycoconjugate e.g., of a glycoprotein, glycolipid, proteoglycan, etc.
  • free glycans including glycans that have been cleaved or otherwise released from a glycoconjugate.
  • a "high mannose glycan” refers to a glycan that includes at least 3 mannose sugar residues and that terminates in a mannose on a non-reducing end of the glycan. In some instances, a "high mannose glycan” includes at least 4, 5, 6, 7, 8, 9, 10, 11, or 12 mannose sugar residues.
  • sialylated glycan refers to a glycan that includes at least 1 sialic acid. In some instances, a sialylated glycan includes at least 1, 2, 3, or 4 sialic acids.
  • a sialylated glycan is a monosialylated glycan (e.g., a branched glycan monosialylated on an ⁇ 1-3 arm of the branched glycan (e.g., with a NeuAc- ⁇ 2,6-Gal terminal linkage)), and/or a disialylated glycan (e.g., a branched glycan sialylated on both an ⁇ 1-3 arm and an ⁇ 1-6 arm of the branched glycan).
  • a monosialylated glycan e.g., a branched glycan monosialylated on an ⁇ 1-3 arm of the branched glycan (e.g., with a NeuAc- ⁇ 2,6-Gal terminal linkage)
  • a disialylated glycan e.g., a branched glycan sialylated on
  • glycoprotein preparation refers to a set of individual glycoprotein molecules, each of which comprises a polypeptide having a particular amino acid sequence (which amino acid sequence includes at least one glycosylation site) and at least one glycan covalently attached to the at least one glycosylation site.
  • Individual molecules of a particular glycoprotein within a glycoprotein preparation typically have identical amino acid sequences but may differ in the occupancy of the at least one glycosylation sites and/or in the identity of the glycans linked to the at least one of the glycosylation sites. That is, a glycoprotein preparation may contain only a single glycoform of a particular glycoprotein, but more typically contains a plurality of glycoforms. Different preparations of the same glycoprotein may differ in the identity of glycoforms present (e.g., a glycoform that is present in one preparation may be absent from another) and/or in the relative amounts of different glycoforms.
  • glycoform is used herein to refer to a particular form of a glycoprotein. That is, when a glycoprotein includes a particular polypeptide that has the potential to be linked to different glycans or sets of glycans, then each different version of the glycoprotein (i.e., where the polypeptide is linked to a particular glycan or set of glycans) is referred to as a "glycoform".
  • Reference glycoprotein refers to a glycoprotein having substantially the same amino acid sequence as (e.g., having about 95-100% identical amino acids of) a glycoprotein described herein, e.g., a glycoprotein to which it is compared.
  • a reference glycoprotein is a therapeutic glycoprotein described herein, e.g., an FDA approved therapeutic glycoprotein.
  • an "N-glycosylation site of an Fc region” refers to an amino acid residue within an Fc region to which a glycan is N-linked.
  • Target value refers to a predetermined level of one or more particular glycans, such as high mannose glycans, fucosylated glycans, and/or sialylated glycans.
  • a target value is an absolute value. In some instances, a target value is a relative value.
  • a target value is a level of one or more particular glycans, such as high mannose glycans (e.g., HM3, HM4, HM5, HM6, HM7, HM8, HM9, or combinations), fucosylated glycans (e.g., G0F, G1F, G2F, or combinations), and/or sialylated glycans (e.g., monosialylated, disialylated, or combinations), in a reference glycoprotein product or described in a specification or master batch record for a pharmaceutical product.
  • glycans such as high mannose glycans (e.g., HM3, HM4, HM5, HM6, HM7, HM8, HM9, or combinations), fucosylated glycans (e.g., G0F, G1F, G2F, or combinations), and/or sialylated glycans (e.g., monosialy
  • a target value refers to an absolute level of (e.g., number of moles of) one or more glycans (e.g., high mannose glycans (e.g., one or more species of high mannose glycans), fucosylated glycans (e.g., one or more species of fucosylated glycans), and/or sialylated glycans (e.g., one or more species of sialylated glycans) in a glycoprotein preparation.
  • glycans e.g., high mannose glycans (e.g., one or more species of high mannose glycans), fucosylated glycans (e.g., one or more species of fucosylated glycans), and/or sialylated glycans (e.g., one or more species of sialylated glycans) in a glycoprotein preparation.
  • a target value refers to a level of one or more glycans (e.g., high mannose glycans (e.g., one or more species of high mannose glycans), fucosylated glycans (e.g., one or more species of fucosylated glycans), and/or sialylated glycans (e.g., one or more species of sialylated glycans) in a glycoprotein preparation relative to total level of glycans in the glycoprotein preparation.
  • glycans e.g., high mannose glycans (e.g., one or more species of high mannose glycans), fucosylated glycans (e.g., one or more species of fucosylated glycans), and/or sialylated glycans (e.g., one or more species of sialylated glycans) in a glycoprotein preparation relative to
  • a target value is expressed as a "percent", which refers to the number of moles of one or more glycans (e.g., Fc glycans) relative to total moles of glycans (e.g., Fc glycans) in a glycoprotein preparation. In some instances, “percent” refers to the number of moles of one or more PNGase F-released Fc glycans relative to total moles of PNGase F-released Fc glycans detected.
  • Any host cell that can be used to express a polypeptide of interest can be used in the methods described herein.
  • the cells can be genetically engineered to contain a recombinant nucleic acid sequence, e.g., a gene, that encodes a polypeptide of interest (e.g., an antibody).
  • useful cells can express a recombinant polypeptide.
  • Recombinant expression of a gene encoding a polypeptide can include construction of an expression vector containing a polynucleotide that encodes the polypeptide. Once a polynucleotide has been obtained, a vector for the production of the polypeptide can be produced by recombinant DNA technology using techniques known in the art.
  • Known methods can be used to construct expression vectors containing polypeptide coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
  • An expression vector can be transferred to a host cell by conventional techniques, and the transfected cells can then be cultured by conventional techniques, modified in accordance with the present disclosure, to produce a recombinant polypeptide.
  • a variety of host expression vector systems can be used (see, e.g., U.S. Pat. No. 5,807,715 ).
  • Such host-expression systems e.g., genetically engineered host expression systems
  • polypeptides e.g., antibodies
  • Such host expression systems include, but are not limited to, yeast (e.g., Saccharomyces and Pichia ) transformed with recombinant yeast expression vectors containing polypeptide coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing polypeptide coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing polypeptide coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, NS0, and 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the aden
  • viral-based expression systems can be utilized (see, e.g., Logan et al., 1984, Proc. Natl. Acad. Sci. USA 8:355-359 ).
  • the efficiency of expression can be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see, e.g., Bittner et al., 1987, Methods in Enzymol. 153:516-544 ).
  • a host cell strain can be chosen that modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products.
  • Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the polypeptide (e.g., antibody) expressed.
  • Such cells include, for example, established mammalian cell lines and insect cell lines, animal cells, fungal cells, and yeast cells.
  • Mammalian host cells include, but are not limited to, CHO, Vero, BHK, HeLa, COS, MDCK, HEK-293, NIH-3T3, W138, BT483, Hs578T, HTB2, BT20, T47D, NS0 (a murine myeloma cell line that does not endogenously produce any immunoglobulin chains), CRL7O3O, HsS78Bst cells, PER.C6, SP2/0-Ag14, and hybridoma cells.
  • Additional, nonlimiting examples of animal or mammalian host cells include Chinese hamster ovary cells (CHO), such as CHO-K1 (ATCC CCL-61), DG44 ( Chasin et al., 1986, Som.
  • CHO-K1 Tet-On cell line (Clontech), CHO designated ECACC 85050302 (CAMR, Salisbury, Wiltshire, UK), CHO clone 13 (GEIMG, Genova, IT), CHO clone B (GEIMG, Genova, IT), CHO-K1/SF designated ECACC 93061607 (CAMR, Salisbury, Wiltshire, UK), RR-CHOK1 designated ECACC 92052129 (CAMR, Salisbury, Wiltshire, UK), CHOKlsv ( Edmonds et al., Mol. Biotech.
  • CHO-S Pichler et al., Biotechnol. Bioeng. 108:386-94 (2011 )
  • dihydrofolate reductase negative CHO cells CHO/-DHFR, Urlaub and Chasin, 1980, Proc. Natl. Acad. Sci. USA, 77:4216
  • dp12.CHO cells U.S. Pat. No. 5,721,121
  • monkey kidney CV1 cells transformed by SV40 COS cells, COS-7, ATCC CRL-1651
  • human embryonic kidney cells e.g., 293 cells, or 293 cells subcloned for growth in suspension culture, Graham et al., 1977, J. Gen.
  • human cervical carcinoma cells HELA, ATCC CCL-2
  • canine kidney cells MDCK, ATCC CCL-34
  • human lung cells W138, ATCC CCL-75
  • human hepatoma cells HEP-G2, HB 8065
  • mouse mammary tumor cells MMT 060562, ATCC CCL-51
  • buffalo rat liver cells BRL 3A, ATCC CRL-1442
  • TR1 cells Mather, 1982, Ann. NY Acad. Sci., 383:44-68 ); MCR 5 cells; and FS4 cells.
  • host cells can be engineered to stably express a polypeptide (e.g., antibody).
  • Host cells can be transformed with DNA controlled by appropriate expression control elements known in the art, including promoter, enhancer, sequences, transcription terminators, polyadenylation sites, and selectable markers. Methods commonly known in the art of recombinant DNA technology can be used to select a desired recombinant clone.
  • a cell is genetically engineered to increase level of expression of an endogenous polypeptide, e.g., by increasing transcription of a gene encoding the polypeptide and/or increasing mRNA stability.
  • transcription of a gene encoding a polypeptide is increased by: altering the regulatory sequence of the endogenous gene, e.g., in a somatic cell, e.g., by the addition of a positive regulatory element, such as an enhancer or a DNA-binding site for a transcriptional activator; the deletion of a negative regulatory element, such as a DNA-binding site for a transcriptional repressor; and/or replacement of the endogenous regulatory sequence, or elements therein, with that of another gene, thereby allowing the coding region of the gene to be transcribed more efficiently.
  • a positive regulatory element such as an enhancer or a DNA-binding site for a transcriptional activator
  • a negative regulatory element such as a DNA-binding site for a transcriptional repressor
  • a polypeptide described herein e.g., an antibody described herein
  • it can be purified by any method known in the art for purification, for example, by chromatography (e.g., ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • chromatography e.g., ion exchange, affinity, and sizing column chromatography
  • centrifugation e.g., centrifugation, differential solubility
  • differential solubility e.g., differential solubility
  • an antibody can be isolated and purified by appropriately selecting and combining affinity columns such as Protein A column with chromatography columns, filtration, ultra filtration, salting-out and dialysis procedures (see Antibodies: A Laboratory Manual, Ed Harlow, David Lane, Cold Spring Harbor Laboratory, 1988 ).
  • a polypeptide e.g., an antibody
  • heterologous polypeptide sequences to facilitate purification.
  • Polypeptides having desired sugar chains can be separated with a lectin column by methods known in the art (see, e.g., WO 02/30954 ).
  • polypeptides e.g., antibodies
  • target levels of glycans e.g., high mannose glycans, fucosylated glycans and/or sialylated glycans
  • glycans e.g., high mannose glycans, fucosylated glycans and/or sialylated glycans
  • DMSO e.g., a particular, effective level of DMSO (e.g., during one or more stages of culture).
  • cells genetically engineered to express a polypeptide are cultured (e.g., at one or more stages of culture) in a medium that includes DMSO, e.g., an elevated level of DMSO, to decrease levels of G0F glycans and/or fucosylated glycans in a preparation of the polypeptide expressed by the cells.
  • a level of G0F glycans and/or fucosylated glycans is decreased relative to the corresponding level(s) in a preparation of the polypeptide produced using the same medium that does not include DMSO, e.g., an elevated level of DMSO.
  • the decreased level of G0F glycans and/or fucosylated glycans is lower than the corresponding level(s) by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%, of the corresponding level.
  • cells genetically engineered to express a polypeptide are cultured (e.g., at one or more stages of culture) in a medium that includes DMSO, e.g., an elevated level of DMSO, to increase levels of high mannose glycans, afucosylated glycans, and/or sialylated glycans in a preparation of the polypeptide expressed by the cells.
  • DMSO e.g., an elevated level of DMSO
  • a level of high mannose glycans, afucosylated glycans, and/or sialylated glycans is increased relative to the corresponding level(s) in a preparation of the polypeptide produced using the same medium that does not include DMSO, e.g., an elevated level of DMSO.
  • the increased level of high mannose glycans, afucosylated glycans, and/or sialylated glycans is higher than the corresponding level(s) by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level.
  • cells genetically engineered to express a polypeptide are cultured (e.g., at one or more stages of culture) in a medium that includes a reduced level of DMSO, to increase levels of G0F glycans and/or fucosylated glycans in a preparation of the polypeptide expressed by the cells.
  • a level of G0F glycans and/or fucosylated glycans is increased relative to the corresponding level(s) in a preparation of the polypeptide produced using the same medium that does not include a reduced level of DMSO.
  • the increased level of G0F glycans and/or fucosylated glycans is higher than the corresponding level(s) by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level.
  • an “elevated level of DMSO” means a higher concentration of DMSO than is present in a standard medium, a starting medium, a medium used at one or more stages of culture, and/or in a medium in which a polypeptide is produced. In some instances, DMSO is not present in a standard and/or starting medium, a medium used at one or more other stages of culture, and/or in a medium in which a polypeptide is produced, and an “elevated level" is any amount of DMSO.
  • a medium can include an elevated level of DMSO initially (i.e., at the start of a culture), and/or medium can be supplemented with DMSO to achieve an elevated level of DMSO at a particular time or times (e.g., at one or more stages) during culturing.
  • an elevated level of DMSO is an increase of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, 1000% or more, relative to a level of DMSO in a standard medium, a starting medium, a medium during one or more stages of culture, and/or in a medium in which a polypeptide is produced.
  • an elevated level of DMSO is an increase in level (v/v) of DMSO of at least about 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%, 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, 5%, or more (v/v) DMSO, relative to a level (v/v) of DMSO in a standard medium, a starting medium, a medium during one or more stages of culture, and/or in a medium in which a polypeptide is produced.
  • an elevated level of DMSO is about 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%, 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, 5%, or higher (v/v) DMSO.
  • an elevated level of DMSO is about 0.1% to 5% DMSO, about 0.1% to about 2% DMSO, about 0.1% to about 1% DMSO, about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 0.1% to about 2.5% DMSO, about 2.5% to about 5% DMSO, about 0.1% to about 2% DMSO, about 0.1% to about 3% DMSO, about 0.1% to about 4% DMSO, about 1% to about 5% DMSO, about 2% to about 5% 5% DMSO, or about 3% to about 5% DMSO.
  • a "reduced level of DMSO” means a lower concentration of DMSO than is present in a standard medium, a starting medium, a medium used at one or more stages of culture, and/or in a medium in which a polypeptide is produced.
  • a medium can include a reduced level of DMSO initially (i.e., at the start of a culture), a medium can be diluted at a particular time or times (e.g., at one or more stages) during culturing to reduce the level of DMSO, and/or a medium can be replaced with a medium having a reduced level of DMSO at a particular time or times (e.g., at one or more stages) during culturing.
  • a reduced level of DMSO is 0% (v/v) DMSO or no detectable level of DMSO in a medium.
  • a reduced level of DMSO is a decrease of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%, relative to a level of DMSO in a standard medium, a starting medium, a medium during one or more stages of culture, and/or in a medium in which a polypeptide is produced.
  • a reduced level of DMSO is a decrease in level (v/v) of DMSO of at least about 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%, 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, 5%, or more, (v/v) DMSO, relative to a level (v/v) of DMSO in a standard medium, a starting medium, a medium during one or more stages of culture, and/or in a medium in which a polypeptide is produced.
  • a reduced level of DMSO is less than about 5%, 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3%, 2.9%, 2.8%, 2.7%, 2.6%, 2.5%, 2.4%, 2.3%, 2.2%, 2.1%, 2%, 1.9%, 1.8%, 1.7%, 1.6%, 1.5%, 1.4%, 1.3%, 1.2%, 1.1%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5%, 0.4%, 0.3%, 0.2%, or 0.1%; or is 0% (v/v) DMSO.
  • Cells can be cultured in a variety of cell culture media known in the art, which are modified according to the disclosure to include DMSO as described herein.
  • Cell culture medium is understood by those of skill in the art to refer to a nutrient solution in which cells, such as animal or mammalian cells, are grown.
  • a cell culture medium generally includes one or more of the following components: an energy source (e.g., a carbohydrate such as glucose); amino acids; vitamins; lipids or free fatty acids; and trace elements, e.g., inorganic compounds or naturally occurring elements in the micromolar range.
  • an energy source e.g., a carbohydrate such as glucose
  • amino acids e.g., amino acids
  • vitamins e.g., lipids or free fatty acids
  • trace elements e.g., inorganic compounds or naturally occurring elements in the micromolar range.
  • Cell culture medium can also contain additional components, such as hormones and other growth factors (e.g., insulin, transferrin, epidermal growth factor, serum, and the like); salts (e.g., calcium, magnesium and phosphate); buffers (e.g., HEPES); nucleosides and bases (e.g., adenosine, thymidine, hypoxanthine); antibiotics (e.g., gentamycin); and cell protective agents (e.g., a Pluronic polyol (Pluronic F68)).
  • hormones and other growth factors e.g., insulin, transferrin, epidermal growth factor, serum, and the like
  • salts e.g., calcium, magnesium and phosphate
  • buffers e.g., HEPES
  • nucleosides and bases e.g., adenosine, thymidine, hypoxanthine
  • antibiotics e.g., gentamycin
  • cell protective agents
  • a cell culture medium in addition to an elevated or reduced level of DMSO, includes lysine, cysteine, ammonium, manganese, copper, cobalt, putrescine, a peptone, glucose, galactose, galactosamine, glucosamine, glutamine, a lipid (e.g., cholesterol), and/or dextran sulfate.
  • a culture medium can include about 0.1 g/L to about 30 g/L lysine (e.g., about 1 g/L to about 10 g/L lysine, about 2 g/L to about 8 g/L lysine, about 3 g/L to about 7 g/L lysine, about 4 g/L to about 6 g/L lysine, about 1 g/L lysine, about 2 g/L lysine, about 3 g/L lysine, about 4 g/L lysine, about 5 g/L lysine, about 6 g/L lysine, about 7 g/L lysine, about 8 g/L lysine, about 9 g/L lysine, or about 10 g/L lysine, or more); about 0.1 g/L to about 1 g/L cysteine; about 1 mM to about 50 mM ammonium (e.g.,
  • Media that has been prepared or commercially available can be modified according to the present disclosure for utilization in the methods described herein.
  • Nonlimiting examples of such media include Minimal Essential Medium (MEM, Sigma, St. Louis, Mo.); Ham's F10 Medium (Sigma); Dulbecco's Modified Eagles Medium (DMEM, Sigma); RPM 1-1640 Medium (Sigma); HyClone cell culture medium (HyClone, Logan, Utah); Power CHO2 (Lonza Inc., Allendale, NJ); and chemically-defined (CD) media, which are formulated for particular cell types, e.g., CD-CHO Medium (Invitrogen, Carlsbad, Calif.).
  • Culture medium suitable for particular cells being cultured can be determined by a person of ordinary skill in the art without undue experimentation, and such medium can be altered according to the disclosure.
  • Cell culture conditions suitable for cellular production of polypeptides described herein (e.g., antibodies) are those that are known in the art, such as conditions for batch, continuous, or fed-batch culturing of cells.
  • pH of cell culture medium is generally maintained at about 6.8 to about 7.2.
  • cells are cultured in one or more stages, and cells can be cultured in medium having an elevated or reduced level of DMSO in one or more stages.
  • a culture method can include a first stage (e.g., using a medium having a reduced level of DMSO) and a second stage (e.g., using a medium having an elevated level of DMSO).
  • a culture method can include a first stage (e.g., using a medium having an elevated level of DMSO) and a second stage (e.g., using a medium having a reduced level of DMSO).
  • a culture method includes more than two stages, e.g., 3, 4, 5, 6, or more stages, and any stage can include medium having an elevated level of DMSO or a reduced level of DMSO.
  • the length of culture is not limiting.
  • a culture method can be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more days.
  • a culture method includes at least two stages.
  • a first stage can include culturing cells in medium having a reduced level of DMSO (e.g., for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more days), and a second stage can include culturing cells in medium having an elevated level of DMSO (e.g., for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more days).
  • a reduced level of DMSO e.g., for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more days
  • an elevated level of DMSO e.g., for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more days
  • cells are cultured in an initial medium having a reduced level of DMSO for 5 days, and medium having an elevated of DMSO is added to the culture of cells on day 6.
  • a first stage of culture is a growth stage.
  • a cell culture undergoes a period of exponential cell growth (the log phase) where cells are generally rapidly dividing.
  • cells are cultured under such conditions such that cell growth is maximized.
  • the growth cycle and conditions for maximizing growth of host cells can be determined for a particular host cell by a person of ordinary skill in the art without undue experimentation.
  • cells are maintained in a growth stage for a period of between 1 and 10 days.
  • cells are cultured in a medium having a reduced level of DMSO or an elevated level of DMSO for all or part of a growth stage.
  • a second stage of culture is a production stage.
  • cell growth has plateaued or is maintained at a near constant level.
  • logarithmic cell growth has ended and polypeptide production is increased.
  • a medium can generally be supplemented to support continued polypeptide production and to achieve a desired polypeptide product.
  • cells are maintained in a production stage for a period of between 1 and 10 days.
  • cells are cultured in a medium having a reduced level of DMSO or an elevated level of DMSO for all or part of a production stage.
  • cell culture methods are classified as batch culture, continuous culture, and fed-batch culture. Any of these culture methods can be used to grow cells that produce polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans).
  • polypeptides e.g., antibodies
  • glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans.
  • the medium comprises an elevated level or a reduced level of DMSO at an initial stage of cell culture.
  • polypeptides e.g., antibodies
  • targeted levels of glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • the culture medium comprises about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO.
  • 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2%
  • the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO.
  • polypeptides e.g., antibodies
  • targeted levels of glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • DMSO reduced level of DMSO
  • Cells are cultured in a medium having about 0.5 to 5% DMSO, such as about 5%, 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3%, 2.9%, 2.8%, 2.7%, 2.6%, 2.5%, 2.4%, 2.3%, 2.2%, 2.1%, 2%, 1.9%, 1.8%, 1.7%, 1.6%, 1.5%, 1.4%, 1.3%, 1.2%, 1.1%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5% (v/v) DMSO.
  • DMSO a medium having about 0.5 to 5% DMSO, such as about 5%, 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3%,
  • Continuous culture is a culture method in which a medium is added and discharged continuously during culture.
  • This continuous method includes perfusion culture.
  • level of DMSO in the medium can be adjusted at one or more stages of culture to result in an elevated level or a reduced level of DMSO.
  • culture medium used at a first stage of culture does not include an elevated level or a reduced level of DMSO, but at a particular time point during continuous culture (such as during all or part of a growth stage and/or a production stage), medium added during culture is elevated or reduced in the level of DMSO.
  • polypeptides e.g., antibodies
  • targeted levels of glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • DMSO e.g., during one or more stages of continuous culture.
  • Cells are cultured, during one or more stages of continuous culture, in a medium having about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO.
  • a medium having about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about
  • the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO.
  • cells are cultured, during one or more stages of continuous culture, in a medium having a reduced level of DMSO. In some instances, cells are cultured, during one or more stages of continuous culture, in a medium having about 0.5 to 5% DMSO, such as about 5%, 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3%, 2.9%, 2.8%, 2.7%, 2.6%, 2.5%, 2.4%, 2.3%, 2.2%, 2.1%, 2%, 1.9%, 1.8%, 1.7%, 1.6%, 1.5%, 1.4%, 1.3%, 1.2%, 1.1%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5% (v/v) DMSO.
  • Fed-batch culture is a method between batch culture and continuous culture.
  • a cell culture is fed or supplemented continuously or sequentially during culture, but unlike continuous culture, discharge of culture solution is not carried out during culture.
  • glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • medium added during one or more stages of culture can have an elevated level or a reduced level of DMSO.
  • polypeptides e.g., antibodies
  • targeted levels of glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • medium at one or more stages
  • glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • DMSO in the culture medium is achieved (e.g., by adding or supplementing with DMSO, e.g., at one or more stages).
  • a level of DMSO of about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO, is achieved.
  • polypeptides e.g., antibodies
  • targeted levels of glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • medium at one or more stages
  • glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • about 0.5 to 5% DMSO such as about 5%, 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3%, 2.9%, 2.8%, 2.7%, 2.6%, 2.5%, 2.4%, 2.3%, 2.2%, 2.1%, 2%, 1.9%, 1.8%, 1.7%, 1.6%, 1.5%, 1.4%, 1.3%, 1.2%, 1.1%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5% (v/v) DMSO in the culture medium is achieved (at one or more stages).
  • cell culture can be carried out under conditions for large or small scale production of polypeptides (e.g., antibodies), using culture vessels and/or culture apparatuses that are conventionally employed for animal or mammalian cell culture.
  • tissue culture dishes, T-flasks, shaker flasks, and spinner flasks can be used on a laboratory scale.
  • a fluidized bed bioreactor, a hollow fiber bioreactor, a roller bottle culture, or a stirred tank bioreactor system can be used (e.g., as described in U.S. Pat. Nos. 7,541,164 and 7,332,303 ).
  • levels of one or more glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • a preparation of polypeptides e.g., antibodies
  • adjustment e.g., increasing or decreasing the amount of DMSO in the culture
  • possibly termination of the culture in order, for example, to achieve a target level of polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans).
  • Described herein are therapeutic preparations of polypeptides (e.g., glycoproteins), and methods of making and using such preparations.
  • Glycoproteins include, for example, any of a variety of hematologic agents (including, for instance, erythropoietin, blood-clotting factors, etc.), interferons, colony stimulating factors, antibodies, enzymes, and hormones.
  • hematologic agents including, for instance, erythropoietin, blood-clotting factors, etc.
  • interferons including, for instance, erythropoietin, blood-clotting factors, etc.
  • colony stimulating factors antibodies, enzymes, and hormones.
  • the identity of a particular glycoprotein is not intended to limit the present disclosure, and a therapeutic preparation described herein can include any glycoprotein of interest, e.g., a glycoprotein having an Fc region.
  • a glycoprotein described herein can include a target-binding domain that binds to a target of interest (e.g., binds to an antigen).
  • a glycoprotein such as an antibody, can bind to a transmembrane polypeptide (e.g., receptor) or ligand (e.g., a growth factor).
  • Exemplary molecular targets (e.g., antigens) for glycoproteins described herein include CD proteins such as CD2, CD3, CD4, CD8, CD11, CD19, CD20, CD22, CD25, CD33, CD34, CD40, CD52; members of the ErbB receptor family such as the EGF receptor (EGFR, HER1, ErbB1), HER2 (ErbB2), HER3 (ErbB3) or HER4 (ErbB4) receptor; macrophage receptors such as CRIg; tumor necrosis factors such as TNF ⁇ or TRAIL/Apo-2; cell adhesion molecules such as LFA-1, Mac1, p150,95, VLA-4, ICAM-1, VCAM and ⁇ v ⁇ 3 integrin including either ⁇ or ⁇ subunits thereof (e.g., anti-CD11a, anti-CD 18 or anti-CD11b antibodies); growth factors and receptors such as EGF, FGFR (e.g., FGFR3) and VEGF, EGF, FG
  • molecular targets include Tweak, B7RP-1, proprotein convertase subtilisin/kexin type 9 (PCSK9), sclerostin, c-kit, Tie-2, c-fms, and anti-Ml.
  • therapeutic polypeptide e.g., glycoprotein
  • glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • the targeted levels are the levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) in a reference polypeptide product (e.g., glycoprotein product).
  • Nonlimiting, exemplary reference glycoprotein products can include abatacept (Orencia®, Bristol-Myers Squibb), abciximab (ReoPro®, Roche), adalimumab (Humira®, Bristol-Myers Squibb), aflibercept (Eylea®, Regeneron Pharmaceuticals), alefacept (Amevive®, Astellas Pharma), alemtuzumab (Campath®, Genzyme/Bayer), basiliximab (Simulect®, Novartis), belatacept (Nulojix®, Bristol-Myers Squibb), belimumab (Benlysta®, GlaxoSmithKline), bevacizumab (Avastin®, Roche), canakinumab (Ilaris®, Novartis), brentuximab vedotin (Adcetris®, Seattle Genetics), certolizumab (CIMZIA®, UCB, Brussels, Belgium
  • a level of one or more glycans e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans
  • a level of one or more glycans in a reference polypeptide product is determined by analyzing one or more preparations (e.g., one or more lots) of the reference polypeptide.
  • a level of one or more glycans in a reference polypeptide product is a range of the one or more glycans in two or more preparations of the reference polypeptide (e.g., two or more lots of the reference polypeptide product).
  • a level of one or more glycans is a range (e.g., spanning a lowest level of the one or more glycans to a highest level of the one or more glycans) in two or more lots of the reference polypeptide product.
  • N-linked oligosaccharide chains are added to a protein in the lumen of the endoplasmic reticulum (see Molecular Biology of the Cell, Garland Publishing, Inc. (Alberts et al., 1994 )).
  • an initial oligosaccharide typically 14-sugar
  • an asparagine residue contained within the target consensus sequence of Asn-X-Ser/Thr, where X may be any amino acid except proline.
  • the structure of this initial oligosaccharide is common to most eukaryotes, and contains 3 glucose, 9 mannose, and 2 N-acetylglucosamine residues.
  • This initial oligosaccharide chain can be trimmed by specific glycosidase enzymes in the endoplasmic reticulum, resulting in a short, branched core oligosaccharide composed of two N-acetylglucosamine and three mannose residues.
  • N-glycans can be subdivided into three distinct groups called “high mannose type”, “hybrid type”, and “complex type”, with a common pentasaccharide core (Man (alpha1,6)-(Man(alpha1,3))-Man(beta1,4)-GlcpNAc(beta 1,4)-GlcpNAc(beta 1,N)-Asn) occurring in all three groups.
  • the glycoprotein After initial processing in the endoplasmic reticulum, the glycoprotein is transported to the Golgi where further processing may take place. If the glycan is transferred to the Golgi before it is completely trimmed to the core pentasaccharide structure, it results in a "high-mannose glycan".
  • one or more monosaccharides units of N-acetylglucosamine may be added to core mannose subunits to form a "complex glycan".
  • Galactose may be added to N-acetylglucosamine subunits, and sialic acid subunits may be added to galactose subunits, resulting in chains that terminate with any of a sialic acid, a galactose or an N-acetylglucosamine residue.
  • a fucose residue may be added to an N-acetylglucosamine residue of the core oligosaccharide.
  • Each of these additions is catalyzed by specific glycosyl transferases, known in the art.
  • Sialic acids are a family of 9-carbon monosaccharides with heterocyclic ring structures. They bear a negative charge via a carboxylic acid group attached to the ring as well as other chemical decorations including N-acetyl and N-glycolyl groups.
  • the two main types of sialyl residues found in glycoproteins produced in mammalian expression systems are N-acetylneuraminic acid (NeuAc) and N-glycolylneuraminic acid (NeuGc). These usually occur as terminal structures attached to galactose (Gal) residues at the non-reducing termini of both N- and O-linked glycans.
  • the glycosidic linkage configurations for these sialyl groups can be either ⁇ 2,3 or ⁇ 2,6.
  • Hybrid glycans comprise characteristics of both high-mannose and complex glycans.
  • one branch of a hybrid glycan may comprise primarily or exclusively mannose residues, while another branch may comprise N-acetylglucosamine, sialic acid, and/or galactose sugars.
  • Antibodies are glycosylated at conserved, N-linked glycosylation sites in the Fc regions of immunoglobulin heavy chains.
  • each heavy chain of an IgG antibody has a single N-linked glycosylation site at Asn297 of the CH2 domain (see Jefferis, Nature Reviews 8:226-234 (2009 )).
  • IgA antibodies have N-linked glycosylation sites within the CH2 and CH3 domains
  • IgE antibodies have N-linked glycosylation sites within the CH3 domain
  • IgM antibodies have N-linked glycosylation sites within the CH1, CH2, CH3, and CH4 domains (see Arnold et al., J. Biol. Chem.
  • Each antibody isotype has a distinct variety of N-linked carbohydrate structures in the constant regions.
  • IgG has a single N-linked biantennary carbohydrate at Asn297 of the CH2 domain in each Fc polypeptide of the Fc region, which also contains the binding sites for C1q and Fc ⁇ R (see Jefferis et al., Immunol. Rev. 163:59-76 (1998 ); and Wright et al., Trends Biotech 15:26-32 (1997 )).
  • the core oligosaccharide normally consists of GlcNAc 2 Man 3 GlcNAc, with differing numbers of outer residues.
  • Variation among individual IgG can occur via attachment of galactose and/or galactose-sialic acid at one or both terminal GlcNAc or via attachment of a third GlcNAc arm (bisecting GlcNAc), and/or attachment of fucose.
  • an IgG antibody consists of two identical light polypeptide chains and two identical heavy polypeptide chains linked together by disulphide bonds.
  • the first domain located at the amino terminus of each chain is variable in amino acid sequence, providing antibody binding specificities found in each individual antibody. These are known as variable heavy (VH) and variable light (VL) regions.
  • the other domains of each chain are relatively invariant in amino acid sequence and are known as constant heavy (CH) and constant light (CL) regions.
  • the light chain includes one variable region (VL) and one constant region (CL).
  • An IgG heavy chain includes a variable region (VH), a first constant region (CH1), a hinge region, a second constant region (CH2), and a third constant region (CH3).
  • VH variable region
  • CH1 first constant region
  • CH2 second constant region
  • CH3 third constant region
  • IgE and IgM antibodies the heavy chain includes an additional constant region (CH4).
  • Antibodies described herein can include, for example, monoclonal antibodies, polyclonal antibodies (e.g., IVIG), multispecific antibodies, human antibodies, humanized antibodies, camelized antibodies, chimeric antibodies, single-chain Fvs (scFv), disulfide-linked Fvs (sdFv), and anti-idiotypic (anti-Id) antibodies, and antigen-binding fragments of any of the above.
  • Antibodies can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass.
  • Fc fragment refers to one or more fragments of an Fc region that retains an Fc function and/or activity described herein, such as binding to an Fc receptor.
  • fragments include fragments that include an N-linked glycosylation site of an Fc region (e.g., an Asn297 of an IgG heavy chain or homologous sites of other antibody isotypes), such as a CH2 domain.
  • antigen binding fragment of an antibody, as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen.
  • binding fragments encompassed within the term "antigen binding fragment" of an antibody include a Fab fragment, a F(ab') 2 fragment, a Fd fragment, a Fv fragment, a scFv fragment, a dAb fragment ( Ward et al., (1989) Nature 341:544-546 ), and an isolated complementarity determining region (CDR).
  • Fab fragment a fragment of Fab
  • F(ab') 2 fragment a Fd fragment
  • Fv fragment fragment
  • a scFv fragment a dAb fragment
  • CDR isolated complementarity determining region
  • Reference glycoproteins described herein e.g., reference antibodies
  • fragments thereof can be produced by any method known in the art for synthesizing glycoproteins (e.g., antibodies) (see, e.g., Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988 ); Brinkman et al., 1995, J. Immunol. Methods 182:41-50 ; WO 92/22324 ; WO 98/46645 ).
  • Chimeric antibodies can be produced using methods described in, e.g., Morrison, 1985, Science 229:1202 , and humanized antibodies by methods described in, e.g., U.S. Pat. No. 6,180,370 .
  • Additional reference antibodies described herein are bispecific antibodies and multivalent antibodies, as described in, e.g., Segal et al., J. Immunol. Methods 248:1-6 (2001 ); and Tutt et al., J. Immunol. 147: 60 (1991 ).
  • glycoproteins or Fc regions or Fc fragments containing one or more N-glycosylation sites thereof
  • heterologous moieties include, but are not limited to, peptides, polypeptides, proteins, fusion proteins, nucleic acid molecules, small molecules, mimetic agents, synthetic drugs, inorganic molecules, and organic molecules.
  • a glycoprotein conjugate is a fusion protein that comprises a peptide, polypeptide, protein scaffold, scFv, dsFv, diabody, Tandab, or an antibody mimetic fused to an Fc region, such as a glycosylated Fc region.
  • a fusion protein can include a linker region connecting an Fc region to a heterologous moiety (see, e.g., Hallewell et al. (1989), J. Biol. Chem. 264, 5260-5268 ; Alfthan et al. (1995), Protein Eng. 8, 725-731 ; Robinson & Sauer (1996)).
  • Exemplary, nonlimiting reference glycoprotein conjugate products include abatacept (Orencia®, Bristol-Myers Squibb), aflibercept (Eylea®, Regeneron Pharmaceuticals), alefacept (Amevive®, Astellas Pharma), belatacept (Nulojix®, Bristol-Myers Squibb), denileukin diftitox (Ontak®, Eisai), etanercept (Enbrel®, Amgen-Pfizer), and rilonacept (Arcalyst®, Regeneron Pharmaceuticals).
  • a glycoprotein conjugate includes an Fc region (or an Fc fragment containing one or more N-glycosylations site thereof) conjugated to a heterologous polypeptide of at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 amino acids.
  • a glycoprotein conjugate includes an Fc region (or an Fc fragment containing one or more N-glycosylation sites thereof) conjugated to one or more marker sequences, such as a peptide to facilitate purification.
  • a particular marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, Calif., 91311).
  • peptide tags useful for purification include, but are not limited to, the hemagglutinin "HA” tag, which corresponds to an epitope derived from the influenza hemagglutinin protein ( Wilson et al., 1984, Cell 37:767 ) and the "Flag" tag.
  • a glycoprotein conjugate includes an Fc region (or Fc fragment containing one or more N-glycosylation sites thereof) conjugated to a diagnostic or detectable agent.
  • Fc region or Fc fragment containing one or more N-glycosylation sites thereof conjugated to a diagnostic or detectable agent.
  • Such fusion proteins can be useful for monitoring or prognosing development or progression of disease or disorder as part of a clinical testing procedure, such as determining efficacy of a particular therapy.
  • Such diagnosis and detection can be accomplished by coupling a glycoprotein to detectable substances including, but not limited to, various enzymes, such as but not limited to horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; prosthetic groups, such as, but not limited to, streptavidin/biotin and avidin/biotin; fluorescent materials, such as, but not limited to, umbelliferone, fluorescein, fluorescein isothiocynate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; luminescent materials, such as, but not limited to, luminol; bioluminescent materials, such as but not limited to, luciferase, luciferin, and aequorin; radioactive materials, such as but not limited to iodine ( 131 I, 125 I, 123 I), carbon ( 14
  • glycans of glycoproteins are analyzed by any available suitable method.
  • glycan structure and composition as described herein are analyzed, for example, by one or more, enzymatic, chromatographic, mass spectrometry (MS), chromatographic followed by MS, electrophoretic methods, electrophoretic methods followed by MS, nuclear magnetic resonance (NMR) methods, and combinations thereof.
  • exemplary enzymatic methods include contacting a glycoprotein preparation with one or more enzymes under conditions and for a time sufficient to release one or more glycan(s) (e.g., one or more exposed glycan(s)).
  • the one or more enzymes include(s) PNGase F.
  • Exemplary chromatographic methods include, but are not limited to, Strong Anion Exchange chromatography using Pulsed Amperometric Detection (SAX-PAD), liquid chromatography (LC), high performance liquid chromatography (HPLC), ultra performance liquid chromatography (UPLC), thin layer chromatography (TLC), amide column chromatography, and combinations thereof.
  • Exemplary mass spectrometry (MS) include, but are not limited to, tandem MS, LC-MS, LC-MS/MS, matrix assisted laser desorption ionisation mass spectrometry (MALDI-MS), Fourier transform mass spectrometry (FTMS), ion mobility separation with mass spectrometry (IMS-MS), electron transfer dissociation (ETD-MS), and combinations thereof.
  • Exemplary electrophoretic methods include, but are not limited to, capillary electrophoresis (CE), CE-MS, gel electrophoresis, agarose gel electrophoresis, acrylamide gel electrophoresis, SDS-polyacrylamide gel electrophoresis (SDS-PAGE) followed by Western blotting using antibodies that recognize specific glycan structures, and combinations thereof.
  • CE capillary electrophoresis
  • CE-MS gel electrophoresis
  • agarose gel electrophoresis agarose gel electrophoresis
  • acrylamide gel electrophoresis acrylamide gel electrophoresis
  • SDS-PAGE SDS-polyacrylamide gel electrophoresis
  • Exemplary nuclear magnetic resonance include, but are not limited to, one-dimensional NMR (1D-NMR), two-dimensional NMR (2D-NMR), correlation spectroscopy magnetic-angle spinning NMR (COSY-NMR), total correlated spectroscopy NMR (TOCSY-NMR), heteronuclear single-quantum coherence NMR (HSQC-NMR), heteronuclear multiple quantum coherence (HMQC-NMR), rotational nuclear overhauser effect spectroscopy NMR (ROESY-NMR), nuclear overhauser effect spectroscopy (NOESY-NMR), and combinations thereof.
  • glycans are analyzed in accordance with the present disclosure using one or more available methods (to give but a few examples, see Anumula, Anal. Biochem., 350(1):1, 2006 ; Klein et al., Anal. Biochem., 179:162, 1989 ; and/or Townsend, R.R. Carbohydrate Analysis" High Performance Liquid Chromatography and Capillary Electrophoresis., Ed. Z.
  • glycans are characterized using one or more of chromatographic methods, electrophoretic methods, nuclear magnetic resonance methods, and combinations thereof. In some instances, glycans are analyzed by labeling with a fluorescent dye and measuring levels of fluorescence.
  • methods for evaluating one or more target protein specific parameters can be performed by one or more of the methods listed in Table 1.
  • Table 1 Exemplary methods of evaluating parameters: Method(s) Relevant literature Parameter C18 UPLC Mass Spec.* Chen and Flynn, Anal.
  • Glycan(s) e.g., N-linked glycan, exposed N-linked glycan, glycan detection, glycan identification, and characterization; site specific glycation; glycoform detection (e.g., parameters 1-7); percent glycosylation; and/or aglycosyl
  • site specific glycation e.g., parameters 1-7
  • percent glycosylation e.g., parameters 1-7
  • percent glycosylation e.g., percent glycosylation
  • aglycosyl Chen and Flynn, J. Am. Soc. Mass Spectrom., 20:1821-1833 (2009 ) Bioanalyzer (reducing/non-reducing)* Forrer et al., Anal.
  • Glycan e.g., N-linked glycan, exposed N-linked glycan
  • LC-MS reducing/non-reducing/alkylated
  • Glycan e.g., N-linked glycan, exposed N-linked glycan
  • Methods include removal (e.g., enzymatic, chemical, and physical) of glycans Goetze et al., Glycobiol., 21:949-959 (2011 ) Xie et al., mAbs, 2:379-394 (2010 ) Anion-exchange chromatography Ahn et al., J. Chrom.
  • the literature recited may pertain to one or more of the methods for determining a parameter described herein.
  • a glycoprotein described herein can be incorporated (e.g., formulated) into a pharmaceutical composition.
  • a pharmaceutical composition is useful as an alternative and/or improved composition for the prevention and/or treatment of one or more diseases relative to a corresponding reference glycoprotein.
  • Pharmaceutical compositions comprising a glycoprotein can be formulated by methods known to those skilled in the art.
  • the pharmaceutical composition can be administered parenterally in the form of an injectable formulation comprising a sterile solution or suspension in water or another pharmaceutically acceptable liquid.
  • the pharmaceutical composition can be formulated by suitably combining the glycoprotein with pharmaceutically acceptable vehicles or media, such as sterile water and physiological saline, vegetable oil, emulsifier, suspension agent, surfactant, stabilizer, flavoring excipient, diluent, vehicle, preservative, binder, followed by mixing in a unit dose form required for generally accepted pharmaceutical practices.
  • pharmaceutically acceptable vehicles or media such as sterile water and physiological saline, vegetable oil, emulsifier, suspension agent, surfactant, stabilizer, flavoring excipient, diluent, vehicle, preservative, binder.
  • a sterile composition for injection can be formulated in accordance with conventional pharmaceutical practices using distilled water for injection as a vehicle.
  • physiological saline or an isotonic solution containing glucose and other supplements such as D-sorbitol, D-mannose, D-mannitol, and sodium chloride may be used as an aqueous solution for injection, optionally in combination with a suitable solubilizing agent, for example, alcohol such as ethanol and polyalcohol such as propylene glycol or polyethylene glycol, and a nonionic surfactant such as polysorbate 80TM, HCO-50 and the like.
  • Nonlimiting examples of oily liquid include sesame oil and soybean oil, and it may be combined with benzyl benzoate or benzyl alcohol as a solubilizing agent.
  • Other items that may be included are a buffer such as a phosphate buffer, or sodium acetate buffer, a soothing agent such as procaine hydrochloride, a stabilizer such as benzyl alcohol or phenol, and an antioxidant.
  • a formulated injection can be packaged in a suitable ampule.
  • a level of one or more glycans described herein can be compared to a predetermined level (e.g., a corresponding level in a reference standard), e.g., to make a decision regarding the composition of the polypeptide preparation, e.g., a decision to classify, select, accept or discard, release or withhold, process into a drug product, ship, move to a different location, formulate, label, package, release into commerce, or sell or offer for sale the polypeptide, e.g., a recombinant antibody.
  • the decision can be to accept, modify or reject a production parameter or parameters used to make the polypeptide, e.g., an antibody.
  • reference standards include a control level (e.g., a polypeptide produced by a different method) or a range or value in a product specification (e.g., an FDA label or Physician's Insert) or quality criterion for a pharmaceutical preparation containing the polypeptide preparation.
  • a control level e.g., a polypeptide produced by a different method
  • a product specification e.g., an FDA label or Physician's Insert
  • methods include taking action (e.g., physical action) in response to the methods disclosed herein.
  • action e.g., physical action
  • a polypeptide preparation is classified, selected, accepted or discarded, released or withheld, processed into a drug product, shipped, moved to a different location, formulated, labeled, packaged, released into commerce, or sold or offered for sale, depending on whether the preselected or target value is met.
  • processing may include formulating (e.g., combining with pharmaceutical excipients), packaging (e.g., in a syringe or vial), labeling, or shipping at least a portion of the polypeptide preparation.
  • processing includes formulating (e.g., combining with pharmaceutical excipients), packaging (e.g., in a syringe or vial), and labeling at least a portion of the preparation as a drug product described herein.
  • processing can include directing and/or contracting another party to process as described herein.
  • a biological activity of a polypeptide preparation is assessed.
  • Biological activity of the preparation can be analyzed by any known method.
  • a binding activity of a polypeptide is assessed (e.g., binding to a receptor).
  • a therapeutic activity of a polypeptide is assessed (e.g., an activity of a polypeptide in decreasing severity or symptom of a disease or condition, or in delaying appearance of a symptom of a disease or condition).
  • a pharmacologic activity of a polypeptide is assessed (e.g., bioavailability, pharmacokinetics, pharmacodynamics).
  • polypeptide e.g., antibody
  • potential adverse activity or toxicity e.g., propensity to cause hypertension, allergic reactions, thrombotic events, seizures, or other adverse events
  • immunogenicity of a polypeptide preparation is assessed, e.g., by determining whether the preparation elicits an antibody response in a subject.
  • Route of administration can be parenteral, for example, administration by injection, transnasal administration, transpulmonary administration, or transcutaneous administration.
  • Administration can be systemic or local by intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection.
  • a suitable means of administration can be selected based on the age and condition of the patient.
  • a single dose of the pharmaceutical composition containing a modified glycoprotein can be selected from a range of 0.001 to 1000 mg/kg of body weight.
  • a dose can be selected in the range of 0.001 to 100000 mg/body weight, but the present disclosure is not limited to such ranges.
  • the dose and method of administration varies depending on the weight, age, condition, and the like of the patient, and can be suitably selected as needed by those skilled in the art.
  • the disclosure is further illustrated by the following example.
  • the example is provided for illustrative purposes only. It is not to be construed as limiting the scope or content of the disclosure in any way.
  • DMSO DMSO-linked immunosorbent assay for human monocyte glycogen
  • CHO cells were grown initially in base media (Power CHO2, Catalog # BE15-771, Lonza Inc., Allendale, NJ) containing soy hydrolysate, Lonza Power Feed A and additional supplements.
  • base media Power CHO2, Catalog # BE15-771, Lonza Inc., Allendale, NJ
  • the cells were fed with Lonza Power Feed A and soy hydrolysate.
  • the cells were fed with Lonza Power Feed A medium and additional supplements with 2.5% DMSO (final concentration) or without DMSO.
  • Cells were fed 2 g/L glucose when the glucose concentration dropped below 2 g/L.
  • Cells were harvested on Days 10, 12, or 14, and the antibodies produced were evaluated for titer and glycan composition.
  • DMSO may be acting, at least in part, by altering the conversion of 3,3,1,0,0 to G0F by GnTII. Further, as shown in Figure 2A , the presence of DMSO had a temporal effect on G0F levels, which declined over days 10, 12, and 14.
  • DMSO increased the total level of high mannose glycans compared to no DMSO. Further, DMSO led to increased levels of high mannose 6, high mannose 3, and high mannose 5 glycans (see Figure 3B ).
  • DMSO also affected the level of fucosylated glycans, resulting in a decreased level of fucosylated glycans ( Figure 4A ), with an increase in afucosylated glycans ( Figure 4B ). Further, DMSO resulted in an increase in sialylated glycans ( Figure 5A ), with a specific increase in the level of 3,5,1,1,0 sialylated glycan ( Figure 5B ; structure shown in Figure 6 ).
  • This Example demonstrates that culturing cells in DMSO can be used to produce polypeptides, expressed by the cells, having particular levels of glycans.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Reproductive Health (AREA)
  • Cell Biology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Description

    FIELD OF THE INVENTION
  • The invention relates generally to cell culture methods. WO2011/061275A1 describes a method of producing glycoproteins in a medium comprising DMSO.
  • BACKGROUND
  • Therapeutic polypeptides are an important class of therapeutic biotechnology products, and therapeutic antibodies (including murine, chimeric, humanized and human antibodies and fragments thereof) account for the majority of therapeutic biologic products.
  • SUMMARY
  • The invention is defined by the claims. Those aspects/instances of the present disclosure which constitute the invention are defined by the claims.
  • In particular, the invention provides a method of producing a recombinant protein preparation, the method comprising:
    1. (a) providing a cell genetically engineered to express a recombinant protein;
    2. (b) culturing the cell in a culture medium comprising 0.5% to 5% DMSO and, optionally, further comprising one or more of glucosamine, lysine, ammonium, copper, putrescine, glucose, a growth factor, a vitamin, a lipid, and a peptone under conditions in which the cell expresses the recombinant protein; and
    3. (c) harvesting a preparation of the recombinant protein produced by the cell that meets a target value, relative to total glycans, of one or more of high mannose glycans, sialylated glycans, 3,3,1,0,0 glycans and fucosylated glycans, wherein
      1. (i) the target value of high mannose glycans is a level at least 20% higher than a level of high mannose glycans, relative to total glycans, in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising 0.5% to 5% DMSO;
      2. (ii) the target value of high mannose glycans is 0.1% to 20% high mannose glycans;
      3. (iii) the target value of fucosylated glycans is 70% to 100% fucosylated glycans;
      4. (iv) the target value of sialylated glycans is a level at least 10% higher than a level of one or more of sialylated glycans, relative to total glycans, in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising 0.5% to 5% DMSO; and/or
      5. (v) the target value of 3,3,1,0,0 glycans is a level at least 10% higher than a level of 3,3,1,0,0 glycans, relative to total glycans, in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising DMSO; and, optionally,
    4. (d) evaluating the level of the one or more of high mannose glycans, sialylated glycans, 3,3,1,0,0 glycans and fucosylated glycans in the recombinant protein preparation; and/or,
    5. (e) formulating the preparation into a drug product.
  • In one aspect, the disclosure features a method of producing a recombinant protein preparation having a target value of one or more of high mannose glycans and fucosylated glycans, the method comprising: (a) providing a cell genetically engineered to express a recombinant protein; (b) culturing the cell in a culture medium comprising (e.g., supplemented with) DMSO under conditions in which the cell expresses the recombinant protein; and (c) harvesting (e.g., purifying or isolating from the cell and/or culture medium) a preparation of the recombinant protein produced by the cell, wherein the preparation has the target value of the one or more of high mannose glycans and fucosylated glycans. In some instances, the culture medium comprises DMSO for a time and in an amount effective to modify (e.g., increase or decrease) one or more of the high mannose glycans and fucosylated glycans of the recombinant protein.
  • The culture medium comprises about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO. In some embodiments the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO.
  • In some instances, the target value is a level of one or more of high mannose glycans and fucosylated glycans in a reference therapeutic product. In some instances, the target value is a level of one or more of high mannose glycans and fucosylated glycans in a reference therapeutic antibody product. In some instances, the target value is a predetermined pharmaceutical product specification or a quality control criterion for a pharmaceutical preparation, e.g., a Certificate of Analysis (CofA), a Certificate of Testing (CofT), or a Master Batch Record. In some instances, the product specification is a product description in an FDA label, a Physician's Insert, a USP monograph, or an EP monograph.
  • In some instances, the reference therapeutic product is selected from the group consisting of: abatacept, abciximab, adalimumab, aflibercept, alefacept, alemtuzumab, basiliximab, bevacizumab, belatacept, certolizumab, cetuximab, daclizumab, eculizumab, efalizumab, entanercept, gemtuzumab, ibritumomab, infliximab, muromonab-CD3, natalizumab, omalizumab, palivizumab; panitumumab, ranibizumab, rilonacept, rituximab, tositumomab, and trastuzumab.
  • The target value is one or more of: (a) at least about 0.1% to about 20% high mannose glycans, e.g., at least about 0.1%, about 0.5%, about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, high mannose glycans; and (b) at least about 70% to 100% fucosylated glycans, e.g., at least about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% fucosylated glycans. High mannose glycans can be, e.g., HM3, HM4, HM5, HM6, HM7, HM8, HM9, or combinations thereof.
  • In some instances, the target value of the one or more of high mannose glycans and fucosylated glycans is higher than a corresponding level in a preparation produced by culturing the cell in the medium not comprising DMSO. In some instances, the target value is higher than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level.
  • In some instances, the method further comprises evaluating the level of one or more of high mannose glycans and fucosylated glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • In another aspect, the disclosure features a method of producing a recombinant protein preparation, the method comprising: (a) providing a target value of one or more of high mannose glycans and fucosylated glycans; (b) providing a cell genetically engineered to express a recombinant protein; (c) culturing the cell in a culture medium comprising (e.g., supplemented with) DMSO under conditions in which the cell expresses the recombinant protein; (d) harvesting a preparation of the recombinant protein produced by the cell; and (e) processing (e.g., one or more of formulating, filling into a container, labeling, packaging) the preparation into a drug product if the preparation meets the target value of the one or more of high mannose glycans and fucosylated glycans. In some instances, the culture medium comprises DMSO for a time and in an amount effective to modify (e.g., increase or decrease) one or more of the high mannose glycans and fucosylated glycans of the recombinant protein.
  • In some instances, the method further comprises evaluating the level of one or more of high mannose glycans and fucosylated glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, or Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • The culture medium comprises about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO. In some embodiments the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO..
  • In some instances, the target value is a level of one or more of high mannose glycans and fucosylated glycans in a reference therapeutic product. In some instances, the target value is a level of one or more of high mannose glycans and fucosylated glycans in a reference therapeutic antibody product. In some instances, the target value is a predetermined pharmaceutical product specification or a quality control criterion for a pharmaceutical preparation, e.g., a Certificate of Analysis (CofA), a Certificate of Testing (CofT), or a Master Batch Record. In some instances, the product specification is a product description in an FDA label, a Physician's Insert, a USP monograph, or an EP monograph.
  • In some instances, the reference therapeutic product is selected from the group consisting of: abatacept, abciximab, adalimumab, aflibercept, alefacept, alemtuzumab, basiliximab, bevacizumab, belatacept, certolizumab, cetuximab, daclizumab, eculizumab, efalizumab, entanercept, gemtuzumab, ibritumomab, infliximab, muromonab-CD3, natalizumab, omalizumab, palivizumab; panitumumab, ranibizumab, rilonacept, rituximab, tositumomab, and trastuzumab.
  • The target value is one or more of: (a) at least about 0.1% to about 20% high mannose glycans, e.g., at least about 0.1%, about 0.5%, about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, high mannose glycans; and (b) at least about 70% to 100% fucosylated glycans, e.g., at least about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% fucosylated glycans. High mannose glycans can be, e.g., HM3, HM4, HM5, HM6, HM7, HM8, HM9, or combinations thereof.
  • In some instances, the target value of the one or more of high mannose glycans and fucosylated glycans is higher than a corresponding level in a preparation produced by culturing the cell in the medium not comprising DMSO. In some instances, the target value is higher than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level.
  • In another aspect, the disclosure features a method of increasing a level of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans in a recombinant protein preparation, the method comprising: (a) providing a cell genetically engineered to express a recombinant protein; (b) culturing the cell in a culture medium comprising (e.g., supplemented with) DMSO under conditions in which the cell expresses the recombinant protein; and (c) harvesting a preparation of the recombinant protein produced by the cell, wherein the preparation has an increased level of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans relative to a corresponding level in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising DMSO. In some instances, the culture medium comprises DMSO for a time and in an amount effective to increase one or more of the high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans of the recombinant protein. In some instances, the method further comprises processing, (e.g., one or more of formulating, filling into a container, labeling, packaging) the preparation into a drug product if the preparation meets a target value of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans.
  • The culture medium comprises about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO. In some embodiments the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO..
  • In some instances, the method further comprises measuring a level of the one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • In some instances, the increased level of the one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans is higher than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level. In some embodiments, the increased level is one or more of: (a) at least about 0.1% to about 20% high mannose glycans, e.g., at least about 0.1%, about 0.5%, about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, or more, high mannose glycans; (b) at least about 70% to 100% fucosylated glycans, e.g., at least about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% fucosylated glycans; and (c) at least about 10% to about 90% sialylated glycans, e.g., at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% sialylated glycans. High mannose glycans can be, e.g., HM3, HM4, HM5, HM6, HM7, HM8, HM9, or combinations thereof.
  • In another aspect, the disclosure features a method of decreasing a level of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans in a recombinant protein preparation, the method comprising: (a) providing a cell genetically engineered to express a recombinant protein; (b) culturing the cell in a culture medium comprising a reduced level of DMSO under conditions in which the cell expresses the recombinant protein; and (c) harvesting a preparation of the recombinant protein produced by the cell, wherein the preparation has a decreased level of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans relative to a corresponding level in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising the reduced level of DMSO. In some instances, the culture medium comprises the reduced level of DMSO for a time and in an amount effective to decrease a level of one or more of the high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans of the recombinant protein. In some instances, the method further comprises processing, (e.g., one or more of formulating, filling into a container, labeling, packaging) the preparation into a drug product if the preparation meets a target value of one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans.
  • The medium comprises about 0.5 to 5% DMSO such as about 5%, 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3%, 2.9%, 2.8%, 2.7%, 2.6%, 2.5%, 2.4%, 2.3%, 2.2%, 2.1%, 2%, 1.9%, 1.8%, 1.7%, 1.6%, 1.5%, 1.4%, 1.3%, 1.2%, 1.1%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5% DMSO.
  • In some instances, the method further comprises measuring a level of the one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • In some instances, the decreased level of the one or more of high mannose glycans, sialylated glycans, and 3,3,1,0,0 glycans is lower than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%, of the corresponding level. In some instances, the decreased level is one or more of: (a) less than about 20% high mannose glycans; (b) less than about 90% fucosylated glycans; and (c) less than about 90% sialylated glycans. High mannose glycans can be, e.g., HM3, HM4, HM5, HM6, HM7, HM8, HM9, or combinations thereof.
  • In another aspect, the disclosure features a method of decreasing a level of one or more fucosylated glycans in a recombinant protein preparation, the method comprising: (a) providing a cell genetically engineered to express a recombinant protein; (b) culturing the cell in a culture medium comprising (e.g., supplemented with) DMSO under conditions in which the cell expresses the recombinant protein; and (c) harvesting a preparation of the recombinant protein produced by the cell, wherein the preparation has a decreased level of one or more fucosylated glycans relative to a corresponding level in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising DMSO. In some instances, the culture medium comprises DMSO for a time and in an amount effective to decrease the level of one or more fucosylated glycans of the recombinant protein. In some instances, the method further comprises processing, (e.g., one or more of formulating, filling into a container, labeling, packaging) the preparation into a drug product if the preparation meets a target value of one or more fucosylated glycans.
  • The culture medium comprises about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO. In some embodiments the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO..
  • In some instances, the method further comprises measuring a level of fucosylated glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • In some instances, the decreased level of the fucosylated glycans is lower than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%, of the corresponding level. In some instances, the decreased level is less than about 100% fucosylated glycans. In some instances, the one or more fucosylated glycans comprise G0F glycans.
  • In another aspect, the disclosure features a method of increasing a level of one or more fucosylated glycans in a recombinant protein preparation, the method comprising: (a) providing a cell genetically engineered to express a recombinant protein; (b) culturing the cell in a culture medium comprising a reduced level of DMSO under conditions in which the cell expresses the recombinant protein; and (c) harvesting a preparation of the recombinant protein produced by the cell, wherein the preparation has an increased level of one or more fucosylated glycans relative to a corresponding level in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising the reduced level of DMSO. In some instances, the culture medium comprises a reduced level of DMSO for a time and in an amount effective to increase the level of one or more fucosylated glycans of the recombinant protein. In some instances, the method further comprises processing, (e.g., one or more of formulating, filling into a container, labeling, packaging) the preparation into a drug product if the preparation meets a target value of one or more fucosylated glycans.
  • The medium comprises about 0.5 to about 5% DMSO
  • In some instances, the method further comprises measuring a level of fucosylated glycans in the recombinant protein preparation. In some instances, the method further comprises recording the level in a print or computer-readable medium, e.g., in a test report, Material Safety Data Sheet (MSDS), batch record, Certificate of Testing (CofT) or Certificate of Analysis (CofA).
  • In some instances, the increased level of the fucosylated glycans is higher than the corresponding level by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level. In some instances, the increased level is at least about 70% to 100% fucosylated glycans, e.g., at least about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100% fucosylated glycans. In some instances, the one or more fucosylated glycans comprise G0F glycans.
  • In some aspects described herein, the culturing step comprises a first stage and a second stage. In some instances, the first stage comprises culturing the cell in a first culture medium comprising a first level of DMSO, and the second stage comprises culturing the cell in a second culture medium comprising a second level of DMSO. In some instances, the first culture medium comprises a reduced level of DMSO relative to the second culture medium and the second culture medium comprises an elevated level of DMSO relative to the first culture medium. In some instances, the first culture medium comprises an elevated level of DMSO (e.g., about 1% to about 5% DMSO), and the second culture medium comprises a reduced level of (e.g., does not comprise) DMSO.
  • In some instances, the first stage comprises culturing the cell in the first culture medium for about 1 to about 8 days, e.g., 1-7, 1-6, 1-5 days. In some instances, the second stage comprises culturing the cell in the second culture medium for about 1 to about 12 days, e.g., 1-10, 1-9, 1-8, 1-7, 1-6 days. In some instances, the first stage is a growth stage. In some instances, the second stage is a production stage.
  • In some aspects described herein, the culture medium further comprises one or more of lysine, cysteine, ammonium, manganese, copper, cobalt, putrescine, a peptone, glucose, galactose, glucosamine, glutamine, a lipid (e.g., cholesterol), and dextran sulfate.
  • In some aspects described herein, the cell is a mammalian cell. In some instances, the mammalian cell is a CHO (e.g., CHO-K1, DG44, CHO-DXB11, CHOK1SV, CHO-S) Vero, BHK, HeLa, COS, MDCK, or HEK-293 cell.
  • In some aspects described herein, the recombinant protein is a recombinant therapeutic product. In some instances, the recombinant protein is a recombinant therapeutic antibody product. In some instances, the recombinant protein is a recombinant therapeutic fusion protein. In some instances, the recombinant protein is abatacept, abciximab, adalimumab, aflibercept, alefacept, alemtuzumab, basiliximab, bevacizumab, belatacept, certolizumab, cetuximab, daclizumab, eculizumab, efalizumab, entanercept, gemtuzumab, ibritumomab, infliximab, muromonab-CD3, natalizumab, omalizumab, palivizumab; panitumumab, ranibizumab, rilonacept, rituximab, tositumomab, and trastuzumab.
  • In some aspects described herein, the conditions in which cells (e.g., mammalian cells) express recombinant proteins comprise (i) a medium having a pH of about 6, about 6.5, about 6.6, about 6.7, about 6.8, about 6.9, about 7, about 7.1, about 7.2, about 7.3, about 7.4, about 7.5, or about 8; (ii) a temperature of about 25°C, about 26°C, about 27°C, about 28°C, about 29°C, about 30°C, about 31°C, about 32°C, about 33°C, about 34°C, about 35°C, about 36°C, about 37 °C, about 38°C, about 39°C, or about 40°C; and/or (iii) a culture volume of about 100 mL, about 200 mL, about 300 mL, about 400 mL, about 500 mL, about 600 mL, about 700 mL, about 800 mL, about 900 mL, about 1 L, about 2 L, about 3 L, about 5 L, about 10 L, about 20 L, about 30 L, about 40 L, about 50 L, about 100 L, about 200 L, about 300 L, about 400 L, about 500 L, about 600 L, about 700 L, about 800 L, about 900 L, about 1000 L, 5,000 L, 10,000 L, 20,000 L, or more.
  • In another aspect, the disclosure features a preparation of a recombinant protein produced using a method described herein.
  • In another aspect, the disclosure features a method of producing a recombinant therapeutic antibody preparation (e.g., abciximab, adalimumab, alemtuzumab, basiliximab, bevacizumab, certolizumab, cetuximab, daclizumab, eculizumab, efalizumab, gemtuzumab, ibritumomab, infliximab, muromonab-CD3, natalizumab, omalizumab, palivizumab, panitumumab, ranibizumab, rituximab, tositumomab, or trastuzumab), the method comprising: (a) providing a target value (e.g., a predetermined pharmaceutical product specification or a quality control criterion for a pharmaceutical preparation, e.g., a Certificate of Analysis (CofA), a Certificate of Testing (CofT), or a Master Batch Record of a reference therapeutic antibody product) of one or more of high mannose glycans (e.g., HM3, HM4, HM5, HM6, HM7, HM8, HM9, or combinations thereof) and fucosylated glycans; (b) providing a CHO cell genetically engineered to express a recombinant antibody; (c) culturing the cell in a culture medium comprising 0.5% to 5% DMSO (e.g., such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO, or about 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO) under conditions in which the cell expresses the recombinant antibody; (d) harvesting (e.g., purifying or isolating from the cell and/or culture medium) a preparation of the recombinant antibody produced by the cell; and (e) formulating (e.g., one or more of formulating, filling into a container, labeling, packaging) the preparation into a drug product if the preparation meets the target value of the one or more of high mannose glycans and fucosylated glycans.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The present teachings described herein will be more fully understood from the following description of various illustrative instances, when read together with the accompanying drawings. It should be understood that the drawings described below are for illustration purposes only and are not intended to limit the scope of the present teachings in any way.
    • FIG. 1 is a schematic illustration of an IgG antibody molecule.
    • FIG. 2A is a graphic representation of G0F glycan levels in preparations of a model antibody from cells grown in medium with or without DMSO. FIG. 2B is a graphic representation of the two G1F isomers (indicated as GIFa and G1Fb), or G2F glycan levels in preparations of a model antibody from cells grown in medium with or without DMSO.
    • FIG. 3A is a graphic representation of total high mannose glycan levels in preparations of a model antibody from cells grown in medium with or without DMSO. FIG. 3B is a graphic representation of high mannose 6 (HM6) glycan level, high mannose 5 (HM5) glycan level, or high mannose 3 and 4 (HM3 and HM4) glycan level in preparations of a model antibody from cells grown in medium with or without DMSO.
    • FIG. 4A is a graphic representation of fucosylated glycan levels in preparations of a model antibody from cells grown in medium with or without DMSO. FIG. 4B is a graphic representation of a fucosylated glycan levels in preparations of a model antibody from cells grown in medium with or without DMSO.
    • FIG. 5A is a graphic representation of total sialylated glycan levels in preparations of a model antibody from cells grown in medium with or without DMSO. FIG. 5B is a graphic representation of 3,5,1,1,0 sialylated glycan levels in preparations of a model antibody from cells grown in medium with or without DMSO.
    • FIG. 6 depicts structural illustrations of 3,3,1,0,0 and 3,5,1,1,0 glycans (triangle = fucose; dark gray circles = mannose; squares = N-acetyl glucosamine; light gray circle (between square and diamond) = galactose; diamond = sialic acid). "2AB" is a dye conjugated to the end of the glycan that is linked to a polypeptide.
    DETAILED DESCRIPTION
  • The inventors have discovered that preparations of polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) can be produced from cells cultured in a medium having dimethylsulfoxide (DMSO), e.g., a particular level of DMSO effective to cause such effect. Surprisingly, culturing the cells in medium comprising DMSO does not affect cell growth, cell viability, or titer. The present disclosure encompasses preparations of polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans), methods of making such polypeptides (e.g., antibodies), and methods of using such polypeptides (e.g., antibodies).
  • Definitions
  • As used herein, "purified" (or "isolated") refers to a nucleic acid sequence (e.g., a polynucleotide) or an amino acid sequence (e.g., a polypeptide) that is substantially free of other components. In some instances, a purified polynucleotide or purified polypeptide is removed or separated from other components present in its natural environment. For example, an isolated polypeptide is one that is separated from other components of a cell in which it was produced (e.g., the endoplasmic reticulum or cytoplasmic proteins and RNA). An isolated polynucleotide is one that is separated from other nuclear components (e.g., histones) and/or from upstream or downstream nucleic acid sequences. An isolated nucleic acid sequence or amino acid sequence can be at least 60% free, or at least 75% free, or at least 90% free, or at least 95% free from other components present in natural environment of the indicated nucleic acid sequence or amino acid sequence.
  • As used herein, "polynucleotide" (or "nucleotide sequence" or "nucleic acid molecule") refers to an oligonucleotide, nucleotide, or polynucleotide, and fragments or portions thereof, and to DNA and RNA of genomic or synthetic origin, which may be single- or double- stranded, and represent the sense or anti-sense strand.
  • As used herein, "polypeptide" (or "amino acid sequence" or "protein") refers to an oligopeptide, peptide, polypeptide, or protein sequence, and fragments or portions thereof, and to naturally occurring or synthetic molecules. "Amino acid sequence" and like terms, such as "polypeptide" or "protein", are not meant to limit the indicated amino acid sequence to the complete, native amino acid sequence associated with the recited protein molecule.
  • The term "pharmaceutically effective amount" or "therapeutically effective amount" refers to an amount (e.g., dose) effective in treating a patient, having a disorder or condition described herein. It is also to be understood herein that a "pharmaceutically effective amount" may be interpreted as an amount giving a desired therapeutic effect, either taken in one dose or in any dosage or route, taken alone or in combination with other therapeutic agents.
  • The term "treatment" or "treating", as used herein, refers to administering a therapy in an amount, manner, and/or mode effective to improve a condition, symptom, or parameter associated with a disorder or condition or to prevent or reduce progression of a disorder or condition, to a degree detectable to one skilled in the art. An effective amount, manner, or mode can vary depending on the subject and may be tailored to the subject.
  • As used herein, the term "antibody" refers to a polypeptide that includes at least one immunoglobulin variable region, e.g., an amino acid sequence that provides an immunoglobulin variable domain or immunoglobulin variable domain sequence. For example, an antibody can include a heavy (H) chain variable region (abbreviated herein as VH), and a light (L) chain variable region (abbreviated herein as VL). In another example, an antibody includes two heavy (H) chain variable regions and two light (L) chain variable regions. The term "antibody" encompasses antigen-binding fragments of antibodies (e.g., single chain antibodies, Fab, F(ab')2, Fd, Fv, and dAb fragments) as well as complete antibodies, e.g., intact immunoglobulins of types IgA, IgG, IgE, IgD, IgM (as well as subtypes thereof). The light chains of the immunoglobulin can be of types kappa or lambda. In some instances, an antibody includes an Fc region. In some instances, an antibody is a therapeutic antibody.
  • As used herein, the term "Fc region" refers to a dimer of two "Fc polypeptides", each "Fc polypeptide" comprising the constant region of an antibody excluding the first constant region immunoglobulin domain. In some instances, an "Fc region" includes two Fc polypeptides linked by one or more disulfide bonds, chemical linkers, or peptide linkers. "Fc polypeptide" refers to the last two constant region immunoglobulin domains of IgA, IgD, and IgG, and the last three constant region immunoglobulin domains of IgE and IgM, and may also include part or all of the flexible hinge N-terminal to these domains. For IgG, "Fc polypeptide" comprises immunoglobulin domains Cgamma2 (Cγ2) and Cgamma3 (Cγ3) and the lower part of the hinge between Cgamma1 (Cγ1) and Cγ2. Although the boundaries of the Fc polypeptide may vary, the human IgG heavy chain Fc polypeptide is usually defined to comprise residues starting at T223 or C226 or P230, to its carboxyl-terminus, wherein the numbering is according to the EU index as in Kabat et al. (1991, NIH Publication 91-3242, National Technical Information Services, Springfield, VA). For IgA, Fc polypeptide comprises immunoglobulin domains Calpha2 (Cα2) and Calpha3 (Cα3) and the lower part of the hinge between Calpha1 (Cα1) and Cα2. An Fc region can be synthetic, recombinant, or generated from natural sources such as IVIG.
  • As used herein, a "glycan" is a sugar. Glycans can be monomers or polymers of sugar residues, but typically contain at least three sugars, and can be linear or branched. A glycan may include natural sugar residues (e.g., glucose, N-acetylglucosamine, N-acetyl neuraminic acid, galactose, mannose, fucose, hexose, arabinose, ribose, xylose, etc.) and/or modified sugars (e.g., 2'-fluororibose, 2'-deoxyribose, phosphomannose, 6'-sulfo N-acetylglucosamine, etc). The term "glycan" includes homo and heteropolymers of sugar residues. The term "glycan" also encompasses a glycan component of a glycoconjugate (e.g., of a glycoprotein, glycolipid, proteoglycan, etc.). The term also encompasses free glycans, including glycans that have been cleaved or otherwise released from a glycoconjugate.
  • As used herein, a "high mannose glycan" refers to a glycan that includes at least 3 mannose sugar residues and that terminates in a mannose on a non-reducing end of the glycan. In some instances, a "high mannose glycan" includes at least 4, 5, 6, 7, 8, 9, 10, 11, or 12 mannose sugar residues.
  • As used herein, a "sialylated glycan" refers to a glycan that includes at least 1 sialic acid. In some instances, a sialylated glycan includes at least 1, 2, 3, or 4 sialic acids. In some instances, a sialylated glycan is a monosialylated glycan (e.g., a branched glycan monosialylated on an α1-3 arm of the branched glycan (e.g., with a NeuAc-α2,6-Gal terminal linkage)), and/or a disialylated glycan (e.g., a branched glycan sialylated on both an α1-3 arm and an α1-6 arm of the branched glycan).
  • As used herein, the term "glycoprotein preparation" refers to a set of individual glycoprotein molecules, each of which comprises a polypeptide having a particular amino acid sequence (which amino acid sequence includes at least one glycosylation site) and at least one glycan covalently attached to the at least one glycosylation site. Individual molecules of a particular glycoprotein within a glycoprotein preparation typically have identical amino acid sequences but may differ in the occupancy of the at least one glycosylation sites and/or in the identity of the glycans linked to the at least one of the glycosylation sites. That is, a glycoprotein preparation may contain only a single glycoform of a particular glycoprotein, but more typically contains a plurality of glycoforms. Different preparations of the same glycoprotein may differ in the identity of glycoforms present (e.g., a glycoform that is present in one preparation may be absent from another) and/or in the relative amounts of different glycoforms.
  • The term "glycoform" is used herein to refer to a particular form of a glycoprotein. That is, when a glycoprotein includes a particular polypeptide that has the potential to be linked to different glycans or sets of glycans, then each different version of the glycoprotein (i.e., where the polypeptide is linked to a particular glycan or set of glycans) is referred to as a "glycoform".
  • "Reference glycoprotein", as used herein, refers to a glycoprotein having substantially the same amino acid sequence as (e.g., having about 95-100% identical amino acids of) a glycoprotein described herein, e.g., a glycoprotein to which it is compared. In some instances, a reference glycoprotein is a therapeutic glycoprotein described herein, e.g., an FDA approved therapeutic glycoprotein.
  • As used herein, an "N-glycosylation site of an Fc region" refers to an amino acid residue within an Fc region to which a glycan is N-linked.
  • "Target value", as used herein, refers to a predetermined level of one or more particular glycans, such as high mannose glycans, fucosylated glycans, and/or sialylated glycans. In some instances, a target value is an absolute value. In some instances, a target value is a relative value. In some instances, a target value is a level of one or more particular glycans, such as high mannose glycans (e.g., HM3, HM4, HM5, HM6, HM7, HM8, HM9, or combinations), fucosylated glycans (e.g., G0F, G1F, G2F, or combinations), and/or sialylated glycans (e.g., monosialylated, disialylated, or combinations), in a reference glycoprotein product or described in a specification or master batch record for a pharmaceutical product.
  • In some instances, a target value refers to an absolute level of (e.g., number of moles of) one or more glycans (e.g., high mannose glycans (e.g., one or more species of high mannose glycans), fucosylated glycans (e.g., one or more species of fucosylated glycans), and/or sialylated glycans (e.g., one or more species of sialylated glycans) in a glycoprotein preparation. In some instances, a target value refers to a level of one or more glycans (e.g., high mannose glycans (e.g., one or more species of high mannose glycans), fucosylated glycans (e.g., one or more species of fucosylated glycans), and/or sialylated glycans (e.g., one or more species of sialylated glycans) in a glycoprotein preparation relative to total level of glycans in the glycoprotein preparation. In some instances, a target value is expressed as a "percent", which refers to the number of moles of one or more glycans (e.g., Fc glycans) relative to total moles of glycans (e.g., Fc glycans) in a glycoprotein preparation. In some instances, "percent" refers to the number of moles of one or more PNGase F-released Fc glycans relative to total moles of PNGase F-released Fc glycans detected.
  • Cells
  • Any host cell that can be used to express a polypeptide of interest (e.g., an antibody) can be used in the methods described herein. The cells can be genetically engineered to contain a recombinant nucleic acid sequence, e.g., a gene, that encodes a polypeptide of interest (e.g., an antibody). For example, useful cells can express a recombinant polypeptide. Recombinant expression of a gene encoding a polypeptide can include construction of an expression vector containing a polynucleotide that encodes the polypeptide. Once a polynucleotide has been obtained, a vector for the production of the polypeptide can be produced by recombinant DNA technology using techniques known in the art. Known methods can be used to construct expression vectors containing polypeptide coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
  • An expression vector can be transferred to a host cell by conventional techniques, and the transfected cells can then be cultured by conventional techniques, modified in accordance with the present disclosure, to produce a recombinant polypeptide. A variety of host expression vector systems can be used (see, e.g., U.S. Pat. No. 5,807,715 ). Such host-expression systems (e.g., genetically engineered host expression systems) can be used to produce polypeptides (e.g., antibodies) and, where desired, subsequently purified. Such host expression systems include, but are not limited to, yeast (e.g., Saccharomyces and Pichia) transformed with recombinant yeast expression vectors containing polypeptide coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing polypeptide coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing polypeptide coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, NS0, and 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter).
  • For expression in mammalian host cells, viral-based expression systems can be utilized (see, e.g., Logan et al., 1984, Proc. Natl. Acad. Sci. USA 8:355-359). The efficiency of expression can be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see, e.g., Bittner et al., 1987, Methods in Enzymol. 153:516-544).
  • In addition, a host cell strain can be chosen that modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the polypeptide (e.g., antibody) expressed. Such cells include, for example, established mammalian cell lines and insect cell lines, animal cells, fungal cells, and yeast cells. Mammalian host cells include, but are not limited to, CHO, Vero, BHK, HeLa, COS, MDCK, HEK-293, NIH-3T3, W138, BT483, Hs578T, HTB2, BT20, T47D, NS0 (a murine myeloma cell line that does not endogenously produce any immunoglobulin chains), CRL7O3O, HsS78Bst cells, PER.C6, SP2/0-Ag14, and hybridoma cells. Additional, nonlimiting examples of animal or mammalian host cells include Chinese hamster ovary cells (CHO), such as CHO-K1 (ATCC CCL-61), DG44 (Chasin et al., 1986, Som. Cell Molec. Genet., 12:555-556; and Kolkekar et al., 1997, Biochem., 36:10901-10909), CHO-DXB11 (G. Urlaub and L.A. Chasin, 1980 Proc. Natl. Acad. Sci., 77: 4216-4220 . L.H. Graf, and L.A. Chasin 1982, Molec. Cell. Biol., 2: 93-96), CHO-K1 Tet-On cell line (Clontech), CHO designated ECACC 85050302 (CAMR, Salisbury, Wiltshire, UK), CHO clone 13 (GEIMG, Genova, IT), CHO clone B (GEIMG, Genova, IT), CHO-K1/SF designated ECACC 93061607 (CAMR, Salisbury, Wiltshire, UK), RR-CHOK1 designated ECACC 92052129 (CAMR, Salisbury, Wiltshire, UK), CHOKlsv (Edmonds et al., Mol. Biotech. 34:179-190 (2006)), CHO-S (Pichler et al., Biotechnol. Bioeng. 108:386-94 (2011)), dihydrofolate reductase negative CHO cells (CHO/-DHFR, Urlaub and Chasin, 1980, Proc. Natl. Acad. Sci. USA, 77:4216), and dp12.CHO cells ( U.S. Pat. No. 5,721,121 ); monkey kidney CV1 cells transformed by SV40 (COS cells, COS-7, ATCC CRL-1651); human embryonic kidney cells (e.g., 293 cells, or 293 cells subcloned for growth in suspension culture, Graham et al., 1977, J. Gen. Virol., 36:59); baby hamster kidney cells (BHK, ATCC CCL-10); monkey kidney cells (CV1, ATCC CCL-70); African green monkey kidney cells (VERO-76, ATCC CRL-1587; VERO, ATCC CCL-81); mouse sertoli cells (TM4, Mather, 1980, Biol. Reprod., 23:243-251); human cervical carcinoma cells (HELA, ATCC CCL-2); canine kidney cells (MDCK, ATCC CCL-34); human lung cells (W138, ATCC CCL-75); human hepatoma cells (HEP-G2, HB 8065); mouse mammary tumor cells (MMT 060562, ATCC CCL-51); buffalo rat liver cells (BRL 3A, ATCC CRL-1442); TR1 cells (Mather, 1982, Ann. NY Acad. Sci., 383:44-68); MCR 5 cells; and FS4 cells.
  • For long-term, high-yield production of recombinant proteins, host cells can be engineered to stably express a polypeptide (e.g., antibody). Host cells can be transformed with DNA controlled by appropriate expression control elements known in the art, including promoter, enhancer, sequences, transcription terminators, polyadenylation sites, and selectable markers. Methods commonly known in the art of recombinant DNA technology can be used to select a desired recombinant clone. In some instances, a cell is genetically engineered to increase level of expression of an endogenous polypeptide, e.g., by increasing transcription of a gene encoding the polypeptide and/or increasing mRNA stability. In some instances, transcription of a gene encoding a polypeptide is increased by: altering the regulatory sequence of the endogenous gene, e.g., in a somatic cell, e.g., by the addition of a positive regulatory element, such as an enhancer or a DNA-binding site for a transcriptional activator; the deletion of a negative regulatory element, such as a DNA-binding site for a transcriptional repressor; and/or replacement of the endogenous regulatory sequence, or elements therein, with that of another gene, thereby allowing the coding region of the gene to be transcribed more efficiently.
  • Once a polypeptide described herein (e.g., an antibody described herein) has been produced by recombinant expression, it can be purified by any method known in the art for purification, for example, by chromatography (e.g., ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins. For example, an antibody can be isolated and purified by appropriately selecting and combining affinity columns such as Protein A column with chromatography columns, filtration, ultra filtration, salting-out and dialysis procedures (see Antibodies: A Laboratory Manual, Ed Harlow, David Lane, Cold Spring Harbor Laboratory, 1988). Further, as described herein, a polypeptide (e.g., an antibody) can be fused to heterologous polypeptide sequences to facilitate purification. Polypeptides having desired sugar chains can be separated with a lectin column by methods known in the art (see, e.g., WO 02/30954 ).
  • In accordance with the present disclosure, there may be employed conventional molecular biology, microbiology, and recombinant DNA techniques within the skill of the art. Such techniques are described in the literature (see, e.g., Sambrook, Fritsch & Maniatis, Molecular Cloning: A Laboratory Manual, Second Edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.; DNA Cloning: A Practical Approach, Volumes I and II (D. N. Glover ed. 1985); Oligonucleotide Synthesis (M. J. Gait ed. 1984); Nucleic Acid Hybridization (B. D. Hames & S. J. Higgins eds. (1985)); Transcription And Translation (B. D. Hames & S. J. Higgins, eds. (1984)); Animal Cell Culture (R. I. Freshney, ed. (1986)); Immobilized Cells and Enzymes (IRL Press, (1986)); B. Perbal, A Practical Guide To Molecular Cloning (1984); F. M. Ausubel et al. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, Inc. (1994).
  • Culture Methods
  • In general, polypeptides (e.g., antibodies) having target levels of glycans (e.g., high mannose glycans, fucosylated glycans and/or sialylated glycans) can be produced by culturing cells in media that contains DMSO, e.g., a particular, effective level of DMSO (e.g., during one or more stages of culture).
  • In some instances, cells genetically engineered to express a polypeptide are cultured (e.g., at one or more stages of culture) in a medium that includes DMSO, e.g., an elevated level of DMSO, to decrease levels of G0F glycans and/or fucosylated glycans in a preparation of the polypeptide expressed by the cells. In some instances, a level of G0F glycans and/or fucosylated glycans is decreased relative to the corresponding level(s) in a preparation of the polypeptide produced using the same medium that does not include DMSO, e.g., an elevated level of DMSO. In some instances, the decreased level of G0F glycans and/or fucosylated glycans is lower than the corresponding level(s) by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100%, of the corresponding level.
  • In some instances, cells genetically engineered to express a polypeptide are cultured (e.g., at one or more stages of culture) in a medium that includes DMSO, e.g., an elevated level of DMSO, to increase levels of high mannose glycans, afucosylated glycans, and/or sialylated glycans in a preparation of the polypeptide expressed by the cells. In some instances, a level of high mannose glycans, afucosylated glycans, and/or sialylated glycans is increased relative to the corresponding level(s) in a preparation of the polypeptide produced using the same medium that does not include DMSO, e.g., an elevated level of DMSO. In some instances, the increased level of high mannose glycans, afucosylated glycans, and/or sialylated glycans is higher than the corresponding level(s) by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level.
  • In some instances, cells genetically engineered to express a polypeptide are cultured (e.g., at one or more stages of culture) in a medium that includes a reduced level of DMSO, to increase levels of G0F glycans and/or fucosylated glycans in a preparation of the polypeptide expressed by the cells. In some instances, a level of G0F glycans and/or fucosylated glycans is increased relative to the corresponding level(s) in a preparation of the polypeptide produced using the same medium that does not include a reduced level of DMSO. In some instances, the increased level of G0F glycans and/or fucosylated glycans is higher than the corresponding level(s) by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, or more, of the corresponding level.
  • As used herein, an "elevated level of DMSO" means a higher concentration of DMSO than is present in a standard medium, a starting medium, a medium used at one or more stages of culture, and/or in a medium in which a polypeptide is produced. In some instances, DMSO is not present in a standard and/or starting medium, a medium used at one or more other stages of culture, and/or in a medium in which a polypeptide is produced, and an "elevated level" is any amount of DMSO. A medium can include an elevated level of DMSO initially (i.e., at the start of a culture), and/or medium can be supplemented with DMSO to achieve an elevated level of DMSO at a particular time or times (e.g., at one or more stages) during culturing.
  • In some instances, an elevated level of DMSO is an increase of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, 350%, 400%, 450%, 500%, 550%, 600%, 650%, 700%, 750%, 800%, 850%, 900%, 950%, 1000% or more, relative to a level of DMSO in a standard medium, a starting medium, a medium during one or more stages of culture, and/or in a medium in which a polypeptide is produced.
  • In some instances, an elevated level of DMSO is an increase in level (v/v) of DMSO of at least about 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%, 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, 5%, or more (v/v) DMSO, relative to a level (v/v) of DMSO in a standard medium, a starting medium, a medium during one or more stages of culture, and/or in a medium in which a polypeptide is produced.
  • In some instances, an elevated level of DMSO is about 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%, 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, 5%, or higher (v/v) DMSO. In some instances, an elevated level of DMSO is about 0.1% to 5% DMSO, about 0.1% to about 2% DMSO, about 0.1% to about 1% DMSO, about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 0.1% to about 2.5% DMSO, about 2.5% to about 5% DMSO, about 0.1% to about 2% DMSO, about 0.1% to about 3% DMSO, about 0.1% to about 4% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO.
  • As used herein, a "reduced level of DMSO" means a lower concentration of DMSO than is present in a standard medium, a starting medium, a medium used at one or more stages of culture, and/or in a medium in which a polypeptide is produced. A medium can include a reduced level of DMSO initially (i.e., at the start of a culture), a medium can be diluted at a particular time or times (e.g., at one or more stages) during culturing to reduce the level of DMSO, and/or a medium can be replaced with a medium having a reduced level of DMSO at a particular time or times (e.g., at one or more stages) during culturing. In some instances, a reduced level of DMSO is 0% (v/v) DMSO or no detectable level of DMSO in a medium.
  • In some instances, a reduced level of DMSO is a decrease of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100%, relative to a level of DMSO in a standard medium, a starting medium, a medium during one or more stages of culture, and/or in a medium in which a polypeptide is produced.
  • In some instances, a reduced level of DMSO is a decrease in level (v/v) of DMSO of at least about 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%, 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, 5%, or more, (v/v) DMSO, relative to a level (v/v) of DMSO in a standard medium, a starting medium, a medium during one or more stages of culture, and/or in a medium in which a polypeptide is produced.
  • In some instances, a reduced level of DMSO is less than about 5%, 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3%, 2.9%, 2.8%, 2.7%, 2.6%, 2.5%, 2.4%, 2.3%, 2.2%, 2.1%, 2%, 1.9%, 1.8%, 1.7%, 1.6%, 1.5%, 1.4%, 1.3%, 1.2%, 1.1%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5%, 0.4%, 0.3%, 0.2%, or 0.1%; or is 0% (v/v) DMSO.
  • Cells can be cultured in a variety of cell culture media known in the art, which are modified according to the disclosure to include DMSO as described herein. Cell culture medium is understood by those of skill in the art to refer to a nutrient solution in which cells, such as animal or mammalian cells, are grown. A cell culture medium generally includes one or more of the following components: an energy source (e.g., a carbohydrate such as glucose); amino acids; vitamins; lipids or free fatty acids; and trace elements, e.g., inorganic compounds or naturally occurring elements in the micromolar range. Cell culture medium can also contain additional components, such as hormones and other growth factors (e.g., insulin, transferrin, epidermal growth factor, serum, and the like); salts (e.g., calcium, magnesium and phosphate); buffers (e.g., HEPES); nucleosides and bases (e.g., adenosine, thymidine, hypoxanthine); antibiotics (e.g., gentamycin); and cell protective agents (e.g., a Pluronic polyol (Pluronic F68)).
  • In some instances, in addition to an elevated or reduced level of DMSO, a cell culture medium includes lysine, cysteine, ammonium, manganese, copper, cobalt, putrescine, a peptone, glucose, galactose, galactosamine, glucosamine, glutamine, a lipid (e.g., cholesterol), and/or dextran sulfate. For example, a culture medium can include about 0.1 g/L to about 30 g/L lysine (e.g., about 1 g/L to about 10 g/L lysine, about 2 g/L to about 8 g/L lysine, about 3 g/L to about 7 g/L lysine, about 4 g/L to about 6 g/L lysine, about 1 g/L lysine, about 2 g/L lysine, about 3 g/L lysine, about 4 g/L lysine, about 5 g/L lysine, about 6 g/L lysine, about 7 g/L lysine, about 8 g/L lysine, about 9 g/L lysine, or about 10 g/L lysine, or more); about 0.1 g/L to about 1 g/L cysteine; about 1 mM to about 50 mM ammonium (e.g., about 10 mM to about 20 mM ammonium, about 11 mM to about 19 mM ammonium, about 12 mM to about 18 mM ammonium, about 13 mM to about 17 mM ammonium, about 14 mM to about 16 mM ammonium, about 10 mM ammonium, about 11 mM ammonium, about 12 mM, ammonium, about 13 mM ammonium, about 14 mM ammonium, about 15 mM ammonium, about 16 mM ammonium, about 17 mM ammonium, about 18 mM ammonium, about 19 mM ammonium, about 20 mM ammonium, or more); about 0.01 mM to about 0.5 mM manganese; about 0.1 µM to about 0.5 mM copper; about 0.1 mg/L to about 30 mg/L cobalt; about 0.01 mg/L to about 5 mg/L putrescine; about 0.1 g/L to about 10 g/L glucose; about 0.5 g/L to about 30 g/L peptone, e.g., a non-animal derived peptone such as soy or cottonseed; about 1 µM to about 1 mM galactosamine; about 0.1 g/L to about 5 g/L glucosamine; and/or about 0.01 g/L to about 0.1 g/L dextran sulfate.
  • Media that has been prepared or commercially available can be modified according to the present disclosure for utilization in the methods described herein. Nonlimiting examples of such media include Minimal Essential Medium (MEM, Sigma, St. Louis, Mo.); Ham's F10 Medium (Sigma); Dulbecco's Modified Eagles Medium (DMEM, Sigma); RPM 1-1640 Medium (Sigma); HyClone cell culture medium (HyClone, Logan, Utah); Power CHO2 (Lonza Inc., Allendale, NJ); and chemically-defined (CD) media, which are formulated for particular cell types, e.g., CD-CHO Medium (Invitrogen, Carlsbad, Calif.). Culture medium suitable for particular cells being cultured can be determined by a person of ordinary skill in the art without undue experimentation, and such medium can be altered according to the disclosure.
  • Cell culture conditions (including pH, O2, CO2, agitation rate and temperature) suitable for cellular production of polypeptides described herein (e.g., antibodies) are those that are known in the art, such as conditions for batch, continuous, or fed-batch culturing of cells. For example, pH of cell culture medium is generally maintained at about 6.8 to about 7.2.
  • In some instances, cells are cultured in one or more stages, and cells can be cultured in medium having an elevated or reduced level of DMSO in one or more stages. For example, a culture method can include a first stage (e.g., using a medium having a reduced level of DMSO) and a second stage (e.g., using a medium having an elevated level of DMSO). In some instances, a culture method can include a first stage (e.g., using a medium having an elevated level of DMSO) and a second stage (e.g., using a medium having a reduced level of DMSO). In some instances, a culture method includes more than two stages, e.g., 3, 4, 5, 6, or more stages, and any stage can include medium having an elevated level of DMSO or a reduced level of DMSO. The length of culture is not limiting. For example, a culture method can be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more days. In some instances, a culture method includes at least two stages. For example, a first stage can include culturing cells in medium having a reduced level of DMSO (e.g., for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more days), and a second stage can include culturing cells in medium having an elevated level of DMSO (e.g., for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more days).
  • In some instances, cells are cultured in an initial medium having a reduced level of DMSO for 5 days, and medium having an elevated of DMSO is added to the culture of cells on day 6.
  • In some instances, a first stage of culture is a growth stage. Generally, during a growth stage, a cell culture undergoes a period of exponential cell growth (the log phase) where cells are generally rapidly dividing. In some instances, cells are cultured under such conditions such that cell growth is maximized. The growth cycle and conditions for maximizing growth of host cells can be determined for a particular host cell by a person of ordinary skill in the art without undue experimentation. In some instances, cells are maintained in a growth stage for a period of between 1 and 10 days. In some instances, cells are cultured in a medium having a reduced level of DMSO or an elevated level of DMSO for all or part of a growth stage.
  • In some instances, a second stage of culture is a production stage. Generally, during a production stage, cell growth has plateaued or is maintained at a near constant level. During a production stage, logarithmic cell growth has ended and polypeptide production is increased. During this period of time, a medium can generally be supplemented to support continued polypeptide production and to achieve a desired polypeptide product. In some instances, cells are maintained in a production stage for a period of between 1 and 10 days. In some instances, cells are cultured in a medium having a reduced level of DMSO or an elevated level of DMSO for all or part of a production stage.
  • In general, cell culture methods are classified as batch culture, continuous culture, and fed-batch culture. Any of these culture methods can be used to grow cells that produce polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans).
  • Batch Culture
  • In batch culture, a small amount of seed culture solution is added to a medium and cells are grown without any addition of a new medium or discharge of culture solution during culture. For the production of polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) using batch culture, the medium comprises an elevated level or a reduced level of DMSO at an initial stage of cell culture.
  • In some instances, polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) are produced by batch culture of cells expressing the polypeptide in a medium having an elevated level of DMSO. The culture medium comprises about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO. In some embodiments the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO.
  • In some instances, polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) are produced by batch culture of cells expressing the polypeptide in a medium having a reduced level of DMSO. Cells are cultured in a medium having about 0.5 to 5% DMSO, such as about 5%, 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3%, 2.9%, 2.8%, 2.7%, 2.6%, 2.5%, 2.4%, 2.3%, 2.2%, 2.1%, 2%, 1.9%, 1.8%, 1.7%, 1.6%, 1.5%, 1.4%, 1.3%, 1.2%, 1.1%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5% (v/v) DMSO.
  • Continuous Culture
  • Continuous culture is a culture method in which a medium is added and discharged continuously during culture. This continuous method includes perfusion culture. For example, in the production of polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) using continuous culture, level of DMSO in the medium can be adjusted at one or more stages of culture to result in an elevated level or a reduced level of DMSO. In certain methods, culture medium used at a first stage of culture does not include an elevated level or a reduced level of DMSO, but at a particular time point during continuous culture (such as during all or part of a growth stage and/or a production stage), medium added during culture is elevated or reduced in the level of DMSO.
  • In some instances, polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) are produced by continuous culture of cells expressing the polypeptide in a medium having an elevated level of DMSO (e.g., during one or more stages of continuous culture). Cells are cultured, during one or more stages of continuous culture, in a medium having about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO. In some embodiments the culture medium comprises 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%. 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, or 5% DMSO.
  • In some instances, cells are cultured, during one or more stages of continuous culture, in a medium having a reduced level of DMSO. In some instances, cells are cultured, during one or more stages of continuous culture, in a medium having about 0.5 to 5% DMSO, such as about 5%, 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3%, 2.9%, 2.8%, 2.7%, 2.6%, 2.5%, 2.4%, 2.3%, 2.2%, 2.1%, 2%, 1.9%, 1.8%, 1.7%, 1.6%, 1.5%, 1.4%, 1.3%, 1.2%, 1.1%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5% (v/v) DMSO.
  • Fed-batch Culture
  • Fed-batch culture is a method between batch culture and continuous culture. In a fed-batch culture, a cell culture is fed or supplemented continuously or sequentially during culture, but unlike continuous culture, discharge of culture solution is not carried out during culture. For example, for the production of polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) using fed-batch culture, medium added during one or more stages of culture can have an elevated level or a reduced level of DMSO.
  • In some instances, polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) are produced by adding medium (at one or more stages) to a fed batch culture of cells expressing the polypeptide sufficient to achieve (at one or more stages) an elevated level of DMSO. In some embodiments, about 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, 2.1%, 2.2%, 2.3%, 2.4%, 2.5%, 2.6%, 2.7%, 2.8%, 2.9%, 3%, 3.1%, 3.2%, 3.3%, 3.4%, 3.5%, 3.6%, 3.7%, 3.8%, 3.9%, 4%, 4.1%, 4.2%, 4.3%, 4.4%, 4.5%, 4.6%, 4.7%, 4.8%, 4.9%, 5%(v/v) DMSO in the culture medium is achieved (e.g., by adding or supplementing with DMSO, e.g., at one or more stages). In some embodiments, a level of DMSO of about 0.5 to about 5% DMSO such as about 0.5% to about 1% DMSO, about 1% to about 1.5% DMSO, about 0.5% to about 1.5% DMSO, about 0.6% to about 1.4% DMSO, about 0.7% to about 1.3% DMSO, about 0.8% to about 1.2% DMSO, about 0.9% to about 1.1% DMSO, about 1% to about 2% DMSO, about 2% to about 3% DMSO, about 3% to about 4% DMSO, about 4% to about 5% DMSO, about 2.5% to about 5% DMSO, about 1% to about 5% DMSO, about 2% to about 5% DMSO, or about 3% to about 5% DMSO, is achieved.
  • In some instances, polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) are produced by adding medium (at one or more stages) to a fed batch culture of cells expressing the polypeptide sufficient to achieve (at one or more stages) a reduced level of DMSO. In some embodiments, about 0.5 to 5% DMSO, such as about 5%, 4.9%, 4.8%, 4.7%, 4.6%, 4.5%, 4.4%, 4.3%, 4.2%, 4.1%, 4%, 3.9%, 3.8%, 3.7%, 3.6%, 3.5%, 3.4%, 3.3%, 3.2%, 3.1%, 3%, 2.9%, 2.8%, 2.7%, 2.6%, 2.5%, 2.4%, 2.3%, 2.2%, 2.1%, 2%, 1.9%, 1.8%, 1.7%, 1.6%, 1.5%, 1.4%, 1.3%, 1.2%, 1.1%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5% (v/v) DMSO in the culture medium is achieved (at one or more stages).
  • According to the present disclosure, cell culture can be carried out under conditions for large or small scale production of polypeptides (e.g., antibodies), using culture vessels and/or culture apparatuses that are conventionally employed for animal or mammalian cell culture. For example, tissue culture dishes, T-flasks, shaker flasks, and spinner flasks can be used on a laboratory scale. For culturing on a larger scale (e.g., 1 L, 10 L, 100 L, 500 L, 5000 L, or more), a fluidized bed bioreactor, a hollow fiber bioreactor, a roller bottle culture, or a stirred tank bioreactor system can be used (e.g., as described in U.S. Pat. Nos. 7,541,164 and 7,332,303 ).
  • In particular methods, levels of one or more glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) in a preparation of polypeptides (e.g., antibodies) are monitored during one or more times (e.g., one or more stages) of cell culture, thereby allowing adjustment (e.g., increasing or decreasing the amount of DMSO in the culture) or possibly termination of the culture in order, for example, to achieve a target level of polypeptides (e.g., antibodies) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans).
  • Polypeptides
  • Described herein are therapeutic preparations of polypeptides (e.g., glycoproteins), and methods of making and using such preparations. Glycoproteins include, for example, any of a variety of hematologic agents (including, for instance, erythropoietin, blood-clotting factors, etc.), interferons, colony stimulating factors, antibodies, enzymes, and hormones. The identity of a particular glycoprotein is not intended to limit the present disclosure, and a therapeutic preparation described herein can include any glycoprotein of interest, e.g., a glycoprotein having an Fc region.
  • A glycoprotein described herein can include a target-binding domain that binds to a target of interest (e.g., binds to an antigen). For example, a glycoprotein, such as an antibody, can bind to a transmembrane polypeptide (e.g., receptor) or ligand (e.g., a growth factor). Exemplary molecular targets (e.g., antigens) for glycoproteins described herein (e.g., antibodies) include CD proteins such as CD2, CD3, CD4, CD8, CD11, CD19, CD20, CD22, CD25, CD33, CD34, CD40, CD52; members of the ErbB receptor family such as the EGF receptor (EGFR, HER1, ErbB1), HER2 (ErbB2), HER3 (ErbB3) or HER4 (ErbB4) receptor; macrophage receptors such as CRIg; tumor necrosis factors such as TNFα or TRAIL/Apo-2; cell adhesion molecules such as LFA-1, Mac1, p150,95, VLA-4, ICAM-1, VCAM and αvβ3 integrin including either α or β subunits thereof (e.g., anti-CD11a, anti-CD 18 or anti-CD11b antibodies); growth factors and receptors such as EGF, FGFR (e.g., FGFR3) and VEGF; IgE; cytokines such as IL1; cytokine receptors such as IL2 receptor; blood group antigens; flk2/flt3 receptor; obesity (OB) receptor; mpl receptor; CTLA-4; protein C; neutropilins; ephrins and receptors; netrins and receptors; slit and receptors; chemokines and chemokine receptors such as CCL5, CCR4, CCR5; amyloid beta; complement factors, such as complement factor D; lipoproteins, such as oxidized LDL (oxLDL); lymphotoxins, such as lymphotoxin alpha (LTa). Other molecular targets include Tweak, B7RP-1, proprotein convertase subtilisin/kexin type 9 (PCSK9), sclerostin, c-kit, Tie-2, c-fms, and anti-Ml.
  • Reference Polypeptides
  • In some instances, described herein are therapeutic polypeptide (e.g., glycoprotein) having targeted levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans), where the targeted levels are the levels of glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) in a reference polypeptide product (e.g., glycoprotein product). Nonlimiting, exemplary reference glycoprotein products can include abatacept (Orencia®, Bristol-Myers Squibb), abciximab (ReoPro®, Roche), adalimumab (Humira®, Bristol-Myers Squibb), aflibercept (Eylea®, Regeneron Pharmaceuticals), alefacept (Amevive®, Astellas Pharma), alemtuzumab (Campath®, Genzyme/Bayer), basiliximab (Simulect®, Novartis), belatacept (Nulojix®, Bristol-Myers Squibb), belimumab (Benlysta®, GlaxoSmithKline), bevacizumab (Avastin®, Roche), canakinumab (Ilaris®, Novartis), brentuximab vedotin (Adcetris®, Seattle Genetics), certolizumab (CIMZIA®, UCB, Brussels, Belgium), cetuximab (Erbitux®, Merck-Serono), daclizumab (Zenapax®, Hoffmann-La Roche), denileukin diftitox (Ontak®, Eisai), denosumab (Prolia®, Amgen; Xgeva®, Amgen), eculizumab (Soliris®, Alexion Pharmaceuticals), efalizumab (Raptiva®, Genentech), etanercept (Enbrel®, Amgen-Pfizer), gemtuzumab (Mylotarg®, Pfizer), golimumab (Simponi®, Janssen), ibritumomab (Zevalin®, Spectrum Pharmaceuticals), infliximab (Remicade®, Centocor), ipilimumab (Yervoy™, Bristol-Myers Squibb), muromonab (Orthoclone OKT3®, Janssen-Cilag), natalizumab (Tysabri®, Biogen Idec, Elan), ofatumumab (Arzerra®, GlaxoSmithKline), omalizumab (Xolair®, Novartis), palivizumab (Synagis®, Medlmmune), panitumumab (Vectibix®, Amgen), ranibizumab (Lucentis®, Genentech), rilonacept (Arcalyst®, Regeneron Pharmaceuticals), rituximab (MabThera®, Roche), tocilizumab (Actemra®, Genentech; RoActemra, Hoffman-La Roche) tositumomab (Bexxar®, GlaxoSmithKline), and trastuzumab (Herceptin®, Roche).
  • In some instances, a level of one or more glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) in a reference polypeptide product is determined by analyzing one or more preparations (e.g., one or more lots) of the reference polypeptide. In some instances, a level of one or more glycans (e.g., high mannose glycans, fucosylated glycans, and/or sialylated glycans) in a reference polypeptide product is a range of the one or more glycans in two or more preparations of the reference polypeptide (e.g., two or more lots of the reference polypeptide product). In some instances, a level of one or more glycans is a range (e.g., spanning a lowest level of the one or more glycans to a highest level of the one or more glycans) in two or more lots of the reference polypeptide product.
  • N-Linked Glycosylation
  • N-linked oligosaccharide chains are added to a protein in the lumen of the endoplasmic reticulum (see Molecular Biology of the Cell, Garland Publishing, Inc. (Alberts et al., 1994)). Specifically, an initial oligosaccharide (typically 14-sugar) is added to the amino group on the side chain of an asparagine residue contained within the target consensus sequence of Asn-X-Ser/Thr, where X may be any amino acid except proline. The structure of this initial oligosaccharide is common to most eukaryotes, and contains 3 glucose, 9 mannose, and 2 N-acetylglucosamine residues. This initial oligosaccharide chain can be trimmed by specific glycosidase enzymes in the endoplasmic reticulum, resulting in a short, branched core oligosaccharide composed of two N-acetylglucosamine and three mannose residues.
  • N-glycans can be subdivided into three distinct groups called "high mannose type", "hybrid type", and "complex type", with a common pentasaccharide core (Man (alpha1,6)-(Man(alpha1,3))-Man(beta1,4)-GlcpNAc(beta 1,4)-GlcpNAc(beta 1,N)-Asn) occurring in all three groups.
  • After initial processing in the endoplasmic reticulum, the glycoprotein is transported to the Golgi where further processing may take place. If the glycan is transferred to the Golgi before it is completely trimmed to the core pentasaccharide structure, it results in a "high-mannose glycan".
  • Additionally or alternatively, one or more monosaccharides units of N-acetylglucosamine may be added to core mannose subunits to form a "complex glycan". Galactose may be added to N-acetylglucosamine subunits, and sialic acid subunits may be added to galactose subunits, resulting in chains that terminate with any of a sialic acid, a galactose or an N-acetylglucosamine residue. Additionally, a fucose residue may be added to an N-acetylglucosamine residue of the core oligosaccharide. Each of these additions is catalyzed by specific glycosyl transferases, known in the art.
  • Sialic acids are a family of 9-carbon monosaccharides with heterocyclic ring structures. They bear a negative charge via a carboxylic acid group attached to the ring as well as other chemical decorations including N-acetyl and N-glycolyl groups. The two main types of sialyl residues found in glycoproteins produced in mammalian expression systems are N-acetylneuraminic acid (NeuAc) and N-glycolylneuraminic acid (NeuGc). These usually occur as terminal structures attached to galactose (Gal) residues at the non-reducing termini of both N- and O-linked glycans. The glycosidic linkage configurations for these sialyl groups can be either α2,3 or α2,6.
  • "Hybrid glycans" comprise characteristics of both high-mannose and complex glycans. For example, one branch of a hybrid glycan may comprise primarily or exclusively mannose residues, while another branch may comprise N-acetylglucosamine, sialic acid, and/or galactose sugars.
  • N-linked Glycosylation in Antibodies
  • Antibodies are glycosylated at conserved, N-linked glycosylation sites in the Fc regions of immunoglobulin heavy chains. For example, each heavy chain of an IgG antibody has a single N-linked glycosylation site at Asn297 of the CH2 domain (see Jefferis, Nature Reviews 8:226-234 (2009)). IgA antibodies have N-linked glycosylation sites within the CH2 and CH3 domains, IgE antibodies have N-linked glycosylation sites within the CH3 domain, and IgM antibodies have N-linked glycosylation sites within the CH1, CH2, CH3, and CH4 domains (see Arnold et al., J. Biol. Chem. 280:29080-29087 (2005); Mattu et al., J. Biol. Chem. 273:2260-2272 (1998); Nettleton et al., Int. Arch. Allergy Immunol. 107:328-329 (1995)).
  • Each antibody isotype has a distinct variety of N-linked carbohydrate structures in the constant regions. For example, IgG has a single N-linked biantennary carbohydrate at Asn297 of the CH2 domain in each Fc polypeptide of the Fc region, which also contains the binding sites for C1q and FcγR (see Jefferis et al., Immunol. Rev. 163:59-76 (1998); and Wright et al., Trends Biotech 15:26-32 (1997)). For human IgG, the core oligosaccharide normally consists of GlcNAc2Man3GlcNAc, with differing numbers of outer residues. Variation among individual IgG can occur via attachment of galactose and/or galactose-sialic acid at one or both terminal GlcNAc or via attachment of a third GlcNAc arm (bisecting GlcNAc), and/or attachment of fucose.
  • Antibodies
  • The basic structure of an IgG antibody is illustrated in Figure 1. As shown in Figure 1, an IgG antibody consists of two identical light polypeptide chains and two identical heavy polypeptide chains linked together by disulphide bonds. The first domain located at the amino terminus of each chain is variable in amino acid sequence, providing antibody binding specificities found in each individual antibody. These are known as variable heavy (VH) and variable light (VL) regions. The other domains of each chain are relatively invariant in amino acid sequence and are known as constant heavy (CH) and constant light (CL) regions. As shown in Figure 1, for an IgG antibody, the light chain includes one variable region (VL) and one constant region (CL). An IgG heavy chain includes a variable region (VH), a first constant region (CH1), a hinge region, a second constant region (CH2), and a third constant region (CH3). In IgE and IgM antibodies, the heavy chain includes an additional constant region (CH4).
  • Antibodies described herein can include, for example, monoclonal antibodies, polyclonal antibodies (e.g., IVIG), multispecific antibodies, human antibodies, humanized antibodies, camelized antibodies, chimeric antibodies, single-chain Fvs (scFv), disulfide-linked Fvs (sdFv), and anti-idiotypic (anti-Id) antibodies, and antigen-binding fragments of any of the above. Antibodies can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass.
  • The term "Fc fragment", as used herein, refers to one or more fragments of an Fc region that retains an Fc function and/or activity described herein, such as binding to an Fc receptor. Examples of such fragments include fragments that include an N-linked glycosylation site of an Fc region (e.g., an Asn297 of an IgG heavy chain or homologous sites of other antibody isotypes), such as a CH2 domain. The term "antigen binding fragment" of an antibody, as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. Examples of binding fragments encompassed within the term "antigen binding fragment" of an antibody include a Fab fragment, a F(ab')2 fragment, a Fd fragment, a Fv fragment, a scFv fragment, a dAb fragment (Ward et al., (1989) Nature 341:544-546), and an isolated complementarity determining region (CDR). These antibody fragments can be obtained using conventional techniques known to those with skill in the art, and fragments can be screened for utility in the same manner as are intact antibodies.
  • Reference glycoproteins described herein (e.g., reference antibodies) or fragments thereof can be produced by any method known in the art for synthesizing glycoproteins (e.g., antibodies) (see, e.g., Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Brinkman et al., 1995, J. Immunol. Methods 182:41-50; WO 92/22324 ; WO 98/46645 ). Chimeric antibodies can be produced using methods described in, e.g., Morrison, 1985, Science 229:1202, and humanized antibodies by methods described in, e.g., U.S. Pat. No. 6,180,370 .
  • Additional reference antibodies described herein are bispecific antibodies and multivalent antibodies, as described in, e.g., Segal et al., J. Immunol. Methods 248:1-6 (2001); and Tutt et al., J. Immunol. 147: 60 (1991).
  • Glycoprotein Conjugates
  • The disclosure includes glycoproteins (or Fc regions or Fc fragments containing one or more N-glycosylation sites thereof) that are conjugated or fused to one or more heterologous moieties. Heterologous moieties include, but are not limited to, peptides, polypeptides, proteins, fusion proteins, nucleic acid molecules, small molecules, mimetic agents, synthetic drugs, inorganic molecules, and organic molecules. In some instances, a glycoprotein conjugate is a fusion protein that comprises a peptide, polypeptide, protein scaffold, scFv, dsFv, diabody, Tandab, or an antibody mimetic fused to an Fc region, such as a glycosylated Fc region. A fusion protein can include a linker region connecting an Fc region to a heterologous moiety (see, e.g., Hallewell et al. (1989), J. Biol. Chem. 264, 5260-5268; Alfthan et al. (1995), Protein Eng. 8, 725-731; Robinson & Sauer (1996)).
  • Exemplary, nonlimiting reference glycoprotein conjugate products include abatacept (Orencia®, Bristol-Myers Squibb), aflibercept (Eylea®, Regeneron Pharmaceuticals), alefacept (Amevive®, Astellas Pharma), belatacept (Nulojix®, Bristol-Myers Squibb), denileukin diftitox (Ontak®, Eisai), etanercept (Enbrel®, Amgen-Pfizer), and rilonacept (Arcalyst®, Regeneron Pharmaceuticals).
  • In some instances, a glycoprotein conjugate includes an Fc region (or an Fc fragment containing one or more N-glycosylations site thereof) conjugated to a heterologous polypeptide of at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 amino acids.
  • In some instances, a glycoprotein conjugate includes an Fc region (or an Fc fragment containing one or more N-glycosylation sites thereof) conjugated to one or more marker sequences, such as a peptide to facilitate purification. A particular marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, Calif., 91311). Other peptide tags useful for purification include, but are not limited to, the hemagglutinin "HA" tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., 1984, Cell 37:767) and the "Flag" tag.
  • In other instances, a glycoprotein conjugate includes an Fc region (or Fc fragment containing one or more N-glycosylation sites thereof) conjugated to a diagnostic or detectable agent. Such fusion proteins can be useful for monitoring or prognosing development or progression of disease or disorder as part of a clinical testing procedure, such as determining efficacy of a particular therapy. Such diagnosis and detection can be accomplished by coupling a glycoprotein to detectable substances including, but not limited to, various enzymes, such as but not limited to horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; prosthetic groups, such as, but not limited to, streptavidin/biotin and avidin/biotin; fluorescent materials, such as, but not limited to, umbelliferone, fluorescein, fluorescein isothiocynate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; luminescent materials, such as, but not limited to, luminol; bioluminescent materials, such as but not limited to, luciferase, luciferin, and aequorin; radioactive materials, such as but not limited to iodine (131I, 125I, 123I), carbon (14C), sulfur (35S), tritium (3H), indium (115In, 113In, 112In, 111In), technetium (99Tc), thallium (201Ti), gallium (68Ga, 67Ga), palladium (103Pd), molybdenum (99Mo), xenon (133Xe), fluorine (18F), 153Sm, 177Lu, 153Gd, 159Gd, 149Pm, 140La, 169Yb, 175Yb, 166Ho, 90Y, 47Sc, 186Re, 188Re, 142Pr, 105Rh, 97Ru, 68Ge, 57Co, 65Zn, 85Sr, 32P, 51Cr, 54Mn, 75Se, 113Sn, and 117Sn; positron emitting metals using various positron emission tomographies, non-radioactive paramagnetic metal ions, and molecules that are radiolabelled or conjugated to specific radioisotopes.
  • Techniques for conjugating therapeutic moieties to antibodies are well known (see, e.g., Arnon et al., "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy", in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56. (Alan R. Liss, Inc. 1985); Hellstrom et al., "Antibodies For Drug Delivery", in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker, Inc. 1987)).
  • Glycan Evaluation
  • In some instances, glycans of glycoproteins are analyzed by any available suitable method. In some instances, glycan structure and composition as described herein are analyzed, for example, by one or more, enzymatic, chromatographic, mass spectrometry (MS), chromatographic followed by MS, electrophoretic methods, electrophoretic methods followed by MS, nuclear magnetic resonance (NMR) methods, and combinations thereof. Exemplary enzymatic methods include contacting a glycoprotein preparation with one or more enzymes under conditions and for a time sufficient to release one or more glycan(s) (e.g., one or more exposed glycan(s)). In some instances, the one or more enzymes include(s) PNGase F. Exemplary chromatographic methods include, but are not limited to, Strong Anion Exchange chromatography using Pulsed Amperometric Detection (SAX-PAD), liquid chromatography (LC), high performance liquid chromatography (HPLC), ultra performance liquid chromatography (UPLC), thin layer chromatography (TLC), amide column chromatography, and combinations thereof. Exemplary mass spectrometry (MS) include, but are not limited to, tandem MS, LC-MS, LC-MS/MS, matrix assisted laser desorption ionisation mass spectrometry (MALDI-MS), Fourier transform mass spectrometry (FTMS), ion mobility separation with mass spectrometry (IMS-MS), electron transfer dissociation (ETD-MS), and combinations thereof. Exemplary electrophoretic methods include, but are not limited to, capillary electrophoresis (CE), CE-MS, gel electrophoresis, agarose gel electrophoresis, acrylamide gel electrophoresis, SDS-polyacrylamide gel electrophoresis (SDS-PAGE) followed by Western blotting using antibodies that recognize specific glycan structures, and combinations thereof. Exemplary nuclear magnetic resonance (NMR) include, but are not limited to, one-dimensional NMR (1D-NMR), two-dimensional NMR (2D-NMR), correlation spectroscopy magnetic-angle spinning NMR (COSY-NMR), total correlated spectroscopy NMR (TOCSY-NMR), heteronuclear single-quantum coherence NMR (HSQC-NMR), heteronuclear multiple quantum coherence (HMQC-NMR), rotational nuclear overhauser effect spectroscopy NMR (ROESY-NMR), nuclear overhauser effect spectroscopy (NOESY-NMR), and combinations thereof.
  • In some instances, techniques described herein may be combined with one or more other technologies for the detection, analysis, and or isolation of glycans or glycoproteins. For example, in certain instances, glycans are analyzed in accordance with the present disclosure using one or more available methods (to give but a few examples, see Anumula, Anal. Biochem., 350(1):1, 2006; Klein et al., Anal. Biochem., 179:162, 1989; and/or Townsend, R.R. Carbohydrate Analysis" High Performance Liquid Chromatography and Capillary Electrophoresis., Ed. Z. E1 Rassi, pp 181-209, 1995; WO2008/128216 ; WO2008/128220 ; WO2008/128218 ; WO2008/130926 ; WO2008/128225 ; WO2008/130924 ; WO2008/128221 ; WO2008/128228 ; WO2008/128227 ; WO2008/128230 ; WO2008/128219 ; WO2008/128222 ; WO2010/071817 ; WO2010/071824 ; WO2010/085251 ; WO2011/069056 ; and WO2011/127322 ,). For example, in some instances, glycans are characterized using one or more of chromatographic methods, electrophoretic methods, nuclear magnetic resonance methods, and combinations thereof. In some instances, glycans are analyzed by labeling with a fluorescent dye and measuring levels of fluorescence.
  • In some instances, methods for evaluating one or more target protein specific parameters, e.g., in a glycoprotein preparation, e.g., one or more of the parameters disclosed herein, can be performed by one or more of the methods listed in Table 1. Table 1: Exemplary methods of evaluating parameters:
    Method(s) Relevant literature Parameter
    C18 UPLC Mass Spec.* Chen and Flynn, Anal. Biochem., 370:147-161 (2007) Glycan(s) (e.g., N-linked glycan, exposed N-linked glycan, glycan detection, glycan identification, and characterization; site specific glycation; glycoform detection (e.g., parameters 1-7); percent glycosylation; and/or aglycosyl)
    Chen and Flynn, J. Am. Soc. Mass Spectrom., 20:1821-1833 (2009)
    Bioanalyzer (reducing/non-reducing)* Forrer et al., Anal. Biochem., 334:81-88 (2004) Glycan (e.g., N-linked glycan, exposed N-linked glycan) (including, for example, glycan detection, identification, and characterization; site specific glycation; glycoform detection; percent glycosylation; and/or aglycosyl)
    LC-MS (reducing/non-reducing/alkylated)* Dick et al., Biotechnol. Bioeng., 100:1132-1143 (2008) Glycan (e.g., N-linked glycan, exposed N-linked glycan) (including, for example, glycan detection, identification, and characterization; site specific glycation; glycoform detection; percent glycosylation; and/or aglycosyl)
    * Methods include removal (e.g., enzymatic, chemical, and physical) of glycans Goetze et al., Glycobiol., 21:949-959 (2011)
    Xie et al., mAbs, 2:379-394 (2010)
    Anion-exchange chromatography Ahn et al., J. Chrom. B, 878:403-408 (2010) Sialylated glycan
    1,2-diamino-4,5-methylenedioxybenzene (DMB) labeling method Hokke et al., FEBS Lett., 275:9-14 (1990) Sialic acid
  • The literature recited may pertain to one or more of the methods for determining a parameter described herein.
  • Pharmaceutical Compositions and Administration
  • A glycoprotein described herein can be incorporated (e.g., formulated) into a pharmaceutical composition. Such a pharmaceutical composition is useful as an alternative and/or improved composition for the prevention and/or treatment of one or more diseases relative to a corresponding reference glycoprotein. Pharmaceutical compositions comprising a glycoprotein can be formulated by methods known to those skilled in the art. The pharmaceutical composition can be administered parenterally in the form of an injectable formulation comprising a sterile solution or suspension in water or another pharmaceutically acceptable liquid. For example, the pharmaceutical composition can be formulated by suitably combining the glycoprotein with pharmaceutically acceptable vehicles or media, such as sterile water and physiological saline, vegetable oil, emulsifier, suspension agent, surfactant, stabilizer, flavoring excipient, diluent, vehicle, preservative, binder, followed by mixing in a unit dose form required for generally accepted pharmaceutical practices. The amount of active ingredient included in the pharmaceutical preparations is such that a suitable dose within the designated range is provided.
  • A sterile composition for injection can be formulated in accordance with conventional pharmaceutical practices using distilled water for injection as a vehicle. For example, physiological saline or an isotonic solution containing glucose and other supplements such as D-sorbitol, D-mannose, D-mannitol, and sodium chloride may be used as an aqueous solution for injection, optionally in combination with a suitable solubilizing agent, for example, alcohol such as ethanol and polyalcohol such as propylene glycol or polyethylene glycol, and a nonionic surfactant such as polysorbate 80™, HCO-50 and the like.
  • Nonlimiting examples of oily liquid include sesame oil and soybean oil, and it may be combined with benzyl benzoate or benzyl alcohol as a solubilizing agent. Other items that may be included are a buffer such as a phosphate buffer, or sodium acetate buffer, a soothing agent such as procaine hydrochloride, a stabilizer such as benzyl alcohol or phenol, and an antioxidant. A formulated injection can be packaged in a suitable ampule.
  • In some instances, a level of one or more glycans described herein can be compared to a predetermined level (e.g., a corresponding level in a reference standard), e.g., to make a decision regarding the composition of the polypeptide preparation, e.g., a decision to classify, select, accept or discard, release or withhold, process into a drug product, ship, move to a different location, formulate, label, package, release into commerce, or sell or offer for sale the polypeptide, e.g., a recombinant antibody. In other instances, the decision can be to accept, modify or reject a production parameter or parameters used to make the polypeptide, e.g., an antibody. Particular, nonlimiting examples of reference standards include a control level (e.g., a polypeptide produced by a different method) or a range or value in a product specification (e.g., an FDA label or Physician's Insert) or quality criterion for a pharmaceutical preparation containing the polypeptide preparation.
  • In some instances, methods (i.e., evaluation, identification, and production methods) include taking action (e.g., physical action) in response to the methods disclosed herein. For example, a polypeptide preparation is classified, selected, accepted or discarded, released or withheld, processed into a drug product, shipped, moved to a different location, formulated, labeled, packaged, released into commerce, or sold or offered for sale, depending on whether the preselected or target value is met. In some instances, processing may include formulating (e.g., combining with pharmaceutical excipients), packaging (e.g., in a syringe or vial), labeling, or shipping at least a portion of the polypeptide preparation. In some instances, processing includes formulating (e.g., combining with pharmaceutical excipients), packaging (e.g., in a syringe or vial), and labeling at least a portion of the preparation as a drug product described herein. Processing can include directing and/or contracting another party to process as described herein.
  • In some instances, a biological activity of a polypeptide preparation (e.g., an antibody preparation) is assessed. Biological activity of the preparation can be analyzed by any known method. In some instances, a binding activity of a polypeptide is assessed (e.g., binding to a receptor). In some instances, a therapeutic activity of a polypeptide is assessed (e.g., an activity of a polypeptide in decreasing severity or symptom of a disease or condition, or in delaying appearance of a symptom of a disease or condition). In some instances, a pharmacologic activity of a polypeptide is assessed (e.g., bioavailability, pharmacokinetics, pharmacodynamics). For methods of analyzing bioavailability, pharmacokinetics, and pharmacodynamics of glycoprotein therapeutics, see, e.g., Weiner et al., J. Pharm. Biomed. Anal. 15(5):571-9, 1997; Srinivas et al., J. Pharm. Sci. 85(1):1-4, 1996; and Srinivas et al., Pharm. Res. 14(7):911-6, 1997.
  • The particular biological activity or therapeutic activity that can be tested will vary depending on the particular polypeptide (e.g., antibody). The potential adverse activity or toxicity (e.g., propensity to cause hypertension, allergic reactions, thrombotic events, seizures, or other adverse events) of polypeptide preparations can be analyzed by any available method. In some instances, immunogenicity of a polypeptide preparation is assessed, e.g., by determining whether the preparation elicits an antibody response in a subject.
  • Route of administration can be parenteral, for example, administration by injection, transnasal administration, transpulmonary administration, or transcutaneous administration. Administration can be systemic or local by intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection.
  • A suitable means of administration can be selected based on the age and condition of the patient. A single dose of the pharmaceutical composition containing a modified glycoprotein can be selected from a range of 0.001 to 1000 mg/kg of body weight. On the other hand, a dose can be selected in the range of 0.001 to 100000 mg/body weight, but the present disclosure is not limited to such ranges. The dose and method of administration varies depending on the weight, age, condition, and the like of the patient, and can be suitably selected as needed by those skilled in the art.
  • The materials, methods, and examples are illustrative only and not intended to be limiting. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, suitable methods and materials are described herein.
  • The disclosure is further illustrated by the following example. The example is provided for illustrative purposes only. It is not to be construed as limiting the scope or content of the disclosure in any way.
  • EXAMPLE Effect of DMSO on Glycosylation Methods
  • The effect of DMSO on antibody glycoforms of a model antibody produced by CHO cells was analyzed. CHO cells were grown initially in base media (Power CHO2, Catalog # BE15-771, Lonza Inc., Allendale, NJ) containing soy hydrolysate, Lonza Power Feed A and additional supplements. On Day 3, the cells were fed with Lonza Power Feed A and soy hydrolysate. On Day 6, the cells were fed with Lonza Power Feed A medium and additional supplements with 2.5% DMSO (final concentration) or without DMSO. Cells were fed 2 g/L glucose when the glucose concentration dropped below 2 g/L. Cells were harvested on Days 10, 12, or 14, and the antibodies produced were evaluated for titer and glycan composition.
  • Relative quantitation of each glycoform was performed on glycans enzymatically released from the antibody using PNGase F and labeled with the fluorophore 2-aminobenzamide. The relative quantities were based on the fluorescence intensity of the relevant species relative to the total fluorescence intensity in the chromatogram. All glycoforms eluting at the same retention time were considered together.
  • Results
  • The presence or absence of DMSO in the culture medium had minimal impact on titer or maximum VCD. Cells grown in the presence of DMSO had an average maximum VCD of 8.0 x 106 cells/mL and a titer of 1.02 g/L, whereas cells grown in the absence of DMSO had an average maximum VCD of 8.1 x 106 cells/mL and a titer of 0.92 g/L.
  • As shown in Figure 2A, the level of G0F was decreased in antibodies grown in the presence of DMSO (about 7% decrease on day 14). Surprisingly, as shown in Figure 2B, the levels of GIF and G2F were only slightly impacted by the presence of DMSO (by about 1% or less). This indicates that the presence of DMSO did not shift the G0F/G1F/G2F distribution by altering galactosylation levels. Rather, this finding suggests that G0F was diverted to other species. A concomitant increase was seen in 3,3,1,0,0 glycans (structure shown in Figure 6; data not shown). As 3,3,1,0,0 is converted to G0F by GnTII, without wishing to be bound by theory, DMSO may be acting, at least in part, by altering the conversion of 3,3,1,0,0 to G0F by GnTII. Further, as shown in Figure 2A, the presence of DMSO had a temporal effect on G0F levels, which declined over days 10, 12, and 14.
  • The impact of DMSO on high mannose glycans is shown in Figures 3A and 3B. As shown in Figure 3A, DMSO increased the total level of high mannose glycans compared to no DMSO. Further, DMSO led to increased levels of high mannose 6, high mannose 3, and high mannose 5 glycans (see Figure 3B).
  • DMSO also affected the level of fucosylated glycans, resulting in a decreased level of fucosylated glycans (Figure 4A), with an increase in afucosylated glycans (Figure 4B). Further, DMSO resulted in an increase in sialylated glycans (Figure 5A), with a specific increase in the level of 3,5,1,1,0 sialylated glycan (Figure 5B; structure shown in Figure 6).
  • This Example demonstrates that culturing cells in DMSO can be used to produce polypeptides, expressed by the cells, having particular levels of glycans.

Claims (12)

  1. A method of producing a recombinant protein preparation, the method comprising:
    (a) providing a cell genetically engineered to express a recombinant protein;
    (b) culturing the cell in a culture medium comprising 0.5% to 5% DMSO and, optionally, further comprising one or more of glucosamine, lysine, ammonium, copper, putrescine, glucose, a growth factor, a vitamin, a lipid, and a peptone under conditions in which the cell expresses the recombinant protein; and
    (c) harvesting a preparation of the recombinant protein produced by the cell that meets a target value, relative to total glycans, of one or more of high mannose glycans, sialylated glycans, 3,3,1,0,0 glycans and fucosylated glycans, wherein
    (i) the target value of high mannose glycans is a level at least 20% higher than a level of high mannose glycans, relative to total glycans, in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising 0.5% to 5% DMSO;
    (ii) the target value of high mannose glycans is 0.1% to 20% high mannose glycans;
    (iii) the target value of fucosylated glycans is 70% to 100% fucosylated glycans;
    (iv) the target value of sialylated glycans is a level at least 10% higher than a level of one or more of sialylated glycans, relative to total glycans, in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising 0.5% to 5% DMSO; and/or
    (v) the target value of 3,3,1,0,0 glycans is a level at least 10% higher than a level of 3,3,1,0,0 glycans, relative to total glycans, in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising DMSO; and, optionally,
    (d) evaluating the level of the one or more of high mannose glycans, sialylated glycans, 3,3,1,0,0 glycans and fucosylated glycans in the recombinant protein preparation; and/or,
    (e) formulating the preparation into a drug product.
  2. The method of claim 1, wherein the recombinant protein is a recombinant therapeutic protein and/or a recombinant therapeutic antibody.
  3. The method of claim 1 or 2, wherein the target value is:
    i) a level of one or more of high mannose glycans, sialylated glycans, 3,3,1,0,0 glycans and fucosylated glycans in a reference therapeutic product;
    ii) a level of one or more of high mannose glycans, sialylated glycans, 3,3,1,0,0 glycans and fucosylated glycans in a reference therapeutic antibody product; and/or
    iii) a predetermined pharmaceutical product specification.
  4. The method of claim 3, wherein the reference therapeutic product is selected from the group consisting of: adalimumab, abatacept, abciximab, aflibercept, alefacept, alemtuzumab, basiliximab, bevacizumab, belatacept, certolizumab, cetuximab, daclizumab, eculizumab, efalizumab, entanercept, gemtuzumab, ibritumomab, infliximab, muromonab-CD3, natalizumab, omalizumab, palivizumab; panitumumab, ranibizumab, rilonacept, rituximab, tositumomab, and trastuzumab.
  5. The method of claim 1 further comprising:
    (a) providing a target value, relative to total glycans, of one or more of high mannose glycans, sialylated glycans, 3,3,1,0,0 glycans and fucosylated glycans; and
    (b) formulating the preparation into a drug product if the preparation meets the target value of the one or more of high mannose glycans, sialylated glycans, 3,3,1,0,0 glycans and fucosylated glycans, wherein
    (i) the target value of high mannose glycans is a level at least 20% higher than a level of high mannose glycans, relative to total glycans, in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising 0.5% to 5% DMSO;
    (ii) the target value of high mannose glycans is 0.1% to 20% high mannose glycans;
    (iii) the target value of fucosylated glycans is a level at least 10% lower than a level of fucosylated glycans, relative to total glycans, in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising 0.5% to 5% DMSO; and/or
    (iv) the target value of fucosylated glycans is 70% to 100% fucosylated glycans;
    (v) the target value of sialylated glycans is a level at least 10% higher than a level of one or more of sialylated glycans, relative to total glycans, in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising 0.5% to 5% DMSO; and/or
    (vi) the target value of 3,3,1,0,0 glycans is a level at least 10% higher than a level of 3,3,1,0,0 glycans, relative to total glycans, in a preparation of the recombinant protein produced by culturing the cell in the medium not comprising 0.5% to 5% DMSO.
  6. The method of claim 5, further comprising evaluating the level of the one or more of high mannose glycans, sialylated glycans, 3,3,1,0,0 glycans and fucosylated glycans in the recombinant protein preparation.
  7. The method of any one of claims 1-6, wherein the one or more fucosylated glycans comprise G0F glycans.
  8. The method of any one of claims 1-7, wherein the culturing step comprises a first stage and a second stage, wherein the first stage comprises culturing the cell in the culture medium comprising a first level of DMSO, and the second stage comprises culturing the cell in the culture medium comprising a second level of DMSO; and optionally wherein
    (i) the second level is increased relative to the first level;
    (ii) the first level of DMSO is no DMSO and the second level of DMSO is 1% to 5% DMSO;
    (iii) the first stage comprises culturing the cell in the first level of DMSO for 1 to 8 days;
    (iv) the second stage comprises culturing the cell in the second level of DMSO for 1 to 12 days; and/or
    (v) the second stage comprises adding DMSO to the culture medium.
  9. The method of any one of claims 1-8, wherein the cell is a Chinese Hamster Ovary (CHO) cell or a murine myeloma cell; optionally wherein the cell is a CHO-K1 cell, a CHO-DG44 cell, an NS0 cell, or an SP2/0-Ag14 cell.
  10. The method of any one of claims 1-9, wherein the culture medium comprises 0.5% to 1.5% DMSO.
  11. The method of any one of claims 1-10, wherein the recombinant protein is selected from the group consisting of: adalimumab, abatacept, abciximab, aflibercept, alefacept, alemtuzumab, basiliximab, bevacizumab, belatacept, certolizumab, cetuximab, daclizumab, eculizumab, efalizumab, entanercept, gemtuzumab, ibritumomab, infliximab, muromonab-CD3, natalizumab, omalizumab, palivizumab, panitumumab, ranibizumab, rilonacept, rituximab, tositumomab, and trastuzumab.
  12. The method of any one of claims 1-11, further comprising recording the evaluated level of one or more of high mannose glycans, fucosylated glycans, sialylated glycans, and 3,3,1,0,0 glycans in a batch record for the preparation.
EP14769180.2A 2013-03-14 2014-03-12 Methods of cell culture Active EP2968549B2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13/829,292 US8956830B2 (en) 2013-03-14 2013-03-14 Methods of cell culture
PCT/US2014/023892 WO2014150655A1 (en) 2013-03-14 2014-03-12 Methods of cell culture

Publications (4)

Publication Number Publication Date
EP2968549A1 EP2968549A1 (en) 2016-01-20
EP2968549A4 EP2968549A4 (en) 2016-08-17
EP2968549B1 true EP2968549B1 (en) 2019-05-08
EP2968549B2 EP2968549B2 (en) 2022-11-23

Family

ID=51528764

Family Applications (1)

Application Number Title Priority Date Filing Date
EP14769180.2A Active EP2968549B2 (en) 2013-03-14 2014-03-12 Methods of cell culture

Country Status (5)

Country Link
US (3) US8956830B2 (en)
EP (1) EP2968549B2 (en)
CN (1) CN105073134A (en)
ES (1) ES2732679T3 (en)
WO (1) WO2014150655A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11407813B2 (en) 2019-12-06 2022-08-09 Regeneron Pharmaceuticals, Inc. Anti-VEGF protein compositions and methods for producing the same

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2137655T3 (en) * 2007-04-16 2012-12-31 Momenta Pharmaceuticals Inc Defined glycoprotein products and related methods
EP2702077A2 (en) 2011-04-27 2014-03-05 AbbVie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9475858B2 (en) 2011-07-08 2016-10-25 Momenta Pharmaceuticals, Inc. Cell culture process
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9334319B2 (en) 2012-04-20 2016-05-10 Abbvie Inc. Low acidic species compositions
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
SG11201507230PA (en) 2013-03-12 2015-10-29 Abbvie Inc Human antibodies that bind human tnf-alpha and methods of preparing the same
US9217168B2 (en) 2013-03-14 2015-12-22 Momenta Pharmaceuticals, Inc. Methods of cell culture
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US8956830B2 (en) 2013-03-14 2015-02-17 Momenta Pharmaceuticals, Inc. Methods of cell culture
US9677105B2 (en) * 2013-03-14 2017-06-13 Momenta Pharmaceuticals, Inc. Methods of cell culture
EP2968544A4 (en) * 2013-03-15 2016-10-12 Hoffmann La Roche Cell culture media and methods of antibody production
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US20150139988A1 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
MA41685A (en) * 2014-10-17 2017-08-22 Biogen Ma Inc COPPER SUPPLEMENT FOR THE REGULATION OF GLYCOSYLATION IN A MAMMAL CELL CULTURE PROCESS
ES2784503T3 (en) 2014-12-01 2020-09-28 Amgen Inc Procedure for manipulating the level of glycan content of a glycoprotein
EP3072957B1 (en) * 2015-03-23 2019-07-10 Lonza Ltd Methods for controlling protein glycosylation
JP2018528218A (en) * 2015-09-11 2018-09-27 アンドリアン、ブルース Recombinant glycosylated eculizumab and eculizumab variants
EP3788076A1 (en) * 2018-05-01 2021-03-10 Amgen Inc. Antibodies with modulated glycan profiles
CN114990049B (en) * 2022-04-26 2024-01-16 鼎康(武汉)生物医药有限公司 Method for simultaneously regulating glycoform and charge heterogeneity of cell expression product

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004008100A2 (en) 2002-07-15 2004-01-22 Immunex Corporation Methods and media for controlling sialylation of proteins produced by mammalian cells
WO2008128227A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Defined glycoprotein products and related methods
WO2009027041A1 (en) 2007-08-31 2009-03-05 F. Hoffmann-La Roche Ag Glycosylation profile analysis
WO2010138502A2 (en) 2009-05-26 2010-12-02 Momenta Pharmaceuticals, Inc. Production of glycoproteins
EP2325296A1 (en) 2009-11-20 2011-05-25 LEK Pharmaceuticals d.d. Production of glycoproteins with low N-glycolylneuraminic acid (Neu5Gc) content
WO2013009648A2 (en) 2011-07-08 2013-01-17 Momenta Pharmaceuticals, Inc. Cell culture process
EP2971040A1 (en) 2013-03-14 2016-01-20 Momenta Pharmaceuticals, Inc. Methods of cell culture

Family Cites Families (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE30985E (en) 1978-01-01 1982-06-29 Serum-free cell culture media
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
DE3801236A1 (en) 1988-01-18 1989-07-27 Boehringer Mannheim Gmbh PENTOSANE SULFATE MEDIUM
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5122469A (en) 1990-10-03 1992-06-16 Genentech, Inc. Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
JPH07503124A (en) 1991-06-14 1995-04-06 ゾーマ・コーポレーション Antibody fragments and their complexes produced by microorganisms
US5426699A (en) 1993-10-25 1995-06-20 Antec Corporation Method and apparatus for digitizing a scrambled analog video signal
EP0666312A1 (en) 1994-02-08 1995-08-09 Wolfgang A. Renner Process for the improvement of mammalian cell growth
US5721121A (en) 1995-06-06 1998-02-24 Genentech, Inc. Mammalian cell culture process for producing a tumor necrosis factor receptor immunoglobulin chimeric protein
US7227002B1 (en) 1997-04-14 2007-06-05 Micromet Ag Human antibodies that bind human 17-A1/EpCAM tumor antigen
US5804420A (en) 1997-04-18 1998-09-08 Bayer Corporation Preparation of recombinant Factor VIII in a protein free medium
DE59813187D1 (en) 1997-12-03 2005-12-15 Roche Diagnostics Gmbh PROCESS FOR THE PREPARATION OF POLYPEPTIDES WITH APPROPRIATE GLYCOSILATION
US6528286B1 (en) 1998-05-29 2003-03-04 Genentech, Inc. Mammalian cell culture process for producing glycoproteins
ATE348173T1 (en) 1999-04-26 2007-01-15 Genentech Inc CELL CULTIVATION METHOD FOR GLYCOPROTEINS
CA2424977C (en) 2000-10-06 2008-03-18 Kyowa Hakko Kogyo Co., Ltd. Process for purifying antibody
MXPA05006522A (en) 2002-12-23 2006-02-17 Bristol Myers Squibb Co Product quality enhancement in mammalian cell culture processes for protein production.
EP1575998A4 (en) 2002-12-23 2007-07-25 Bristol Myers Squibb Co Mammalian cell culture processes for protein production
KR20060076781A (en) 2003-09-18 2006-07-04 레이븐 바이오테크놀로지스, 인코퍼레이티드 Cell culture media
CA2609060C (en) 2005-06-03 2014-07-15 Urs Weber Production of recombinant il-18 binding protein
JP2009508476A (en) * 2005-08-31 2009-03-05 セントカー・インコーポレーテツド Host cell lines for the production of antibody constant regions with enhanced effector functions
JP5586235B2 (en) 2007-03-02 2014-09-10 ワイス・エルエルシー Use of copper and glutamate in cell culture for the production of polypeptides
WO2008130924A2 (en) 2007-04-16 2008-10-30 Momenta Pharmaceuticals, Inc. Isotopically-labeled glycans
US8361705B2 (en) 2007-04-16 2013-01-29 Momenta Pharmaceuticals, Inc. Characterization of N-glycans using exoglycosidases
WO2008128218A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Characterization of n-glycan mixtures by nuclear magnetic resonance
US20100151499A1 (en) 2007-04-16 2010-06-17 Momenta Pharmaceuticals, Inc. Proteolytic release of glycans
WO2008128230A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Reference glycoprotein products and related methods
WO2008128225A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Multi-dimensional chromatographic methods for separating n-glycans
US8043863B2 (en) 2007-04-16 2011-10-25 Momenta Pharmaceuticals, Inc. MS methods to evaluate glycans
WO2008128216A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Methods for labeling glycans
RU2475751C2 (en) 2007-04-16 2013-02-20 Момента Фармасьютикалз, Инк. Methods related to cell surface glycosylation
EP2135093B1 (en) 2007-04-16 2015-04-15 Momenta Pharmaceuticals, Inc. Analysis of phosphorylated glycans, glcopeptides or glycoproteins by imac
WO2008128219A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Comparative analysis of protein conformations by using 2d noesy nmr spectra
WO2008154014A2 (en) 2007-06-11 2008-12-18 Amgen Inc. A method for culturing mammalian cells to improve recombinant protein production
CN102076865B (en) 2008-05-02 2016-03-16 西雅图基因公司 The antibody reduced for the manufacture of core fucosylation and the method and composition of antibody derivatives
JP2012513030A (en) 2008-12-19 2012-06-07 モメンタ ファーマシューティカルズ インコーポレイテッド Methods for modified glycans
WO2010071817A2 (en) 2008-12-19 2010-06-24 Momenta Pharmaceuticals, Inc. Characterization of o-linked glycans
MX2011007545A (en) 2009-01-22 2011-10-28 Momenta Pharmaceuticals Inc Galactose-alpha-1, 3-galactose-containing n-glycans in glycoprotein products derived from cho cells.
CA2763164A1 (en) * 2009-06-05 2010-12-09 Momenta Pharmaceuticals, Inc. Methods of modulating fucosylation of glycoproteins
WO2011069056A2 (en) 2009-12-04 2011-06-09 Momenta Pharmaceuticals, Inc. Antennary fucosylation in glycoproteins from cho cells
SG184051A1 (en) 2010-03-25 2012-10-30 Agency Science Tech & Res Method of producing recombinant proteins with mannose- terminated n-glycans
BR112012025645A2 (en) 2010-04-07 2017-12-12 Momenta Pharmaceuticals Inc high mannose glycans.
JP5953502B2 (en) 2010-07-08 2016-07-20 バクスアルタ ゲーエムベーハー Recombinant high molecular weight vWF production method by cell culture
RU2013122843A (en) 2010-10-20 2014-11-27 Морфотек, Инк. Glycoforms of Antibodies Against Folate Alpha Receptor
WO2012125553A2 (en) 2011-03-12 2012-09-20 Momenta Pharmaceuticals, Inc. N-acetylhexosamine-containing n-glycans in glycoprotein products
US9677105B2 (en) * 2013-03-14 2017-06-13 Momenta Pharmaceuticals, Inc. Methods of cell culture
US8956830B2 (en) * 2013-03-14 2015-02-17 Momenta Pharmaceuticals, Inc. Methods of cell culture

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004008100A2 (en) 2002-07-15 2004-01-22 Immunex Corporation Methods and media for controlling sialylation of proteins produced by mammalian cells
WO2008128227A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Defined glycoprotein products and related methods
WO2009027041A1 (en) 2007-08-31 2009-03-05 F. Hoffmann-La Roche Ag Glycosylation profile analysis
WO2010138502A2 (en) 2009-05-26 2010-12-02 Momenta Pharmaceuticals, Inc. Production of glycoproteins
US20120329709A1 (en) 2009-05-26 2012-12-27 Brian Edward Collins Production of glycoproteins
EP2325296A1 (en) 2009-11-20 2011-05-25 LEK Pharmaceuticals d.d. Production of glycoproteins with low N-glycolylneuraminic acid (Neu5Gc) content
WO2011061275A1 (en) 2009-11-20 2011-05-26 Lek Pharmaceuticals D.D. Production of glycoproteins with low n-glycolylneuraminic acid (neu5gc) content
WO2013009648A2 (en) 2011-07-08 2013-01-17 Momenta Pharmaceuticals, Inc. Cell culture process
EP2971040A1 (en) 2013-03-14 2016-01-20 Momenta Pharmaceuticals, Inc. Methods of cell culture

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
"Adv Biochem Engin/Biotechnol", vol. 127, 2011, article PATRIK HOSSLER: "Protein Glycosylation Control in Mammalian Cell Culture: Past Precedents and Contemporary Prospects", pages: 187 - 219, XP055679442
AKIRA MIZOGUCHI ET AL.: "Changes in asparagine-linked sugar chains of human promyelocytic leukemic cells (HL-60) during monocytoid differentiation and myeloid differentiation. Appearance of high mannose-type oligosaccharides in neutral fraction", THE JOURNAL OF BIOLOGICAL CHEMISTRY, 1984, XP055679545
ANDRE N. ET AL.: "Enhancing functional production of G protein-coupled receptors in Pichia pastoris to levels required for structural studies via a single expression screen", PROTEIN SCIENCE, vol. 15, 2006, pages 1115 - 1126, XP055679445
ARUN KUMAR SHUKLA ET AL.: "Dimethylsulphoxide as a tool to increase functional expression of heterologously produced GPCRs in mammalian cells", FEBS LETTERS, 2006, pages 4261 - 4265, XP005553322
J. RODRIGUEZ ET AL.: "Enhanced production of monomeric interferon-[beta] by CHO cells through the control of culture conditions", BIOTECHNOL. PROG., 2005, pages 22 - 30, XP002572995
JAMES N. ARNOLD ET AL.: "The impact of glycosylation on the biological function and structure of human immunoglobulins", ANNU. REV. IMMUNOL., 2007, pages 21 - 50, XP008081152
JIANXIN YE ET AL.: "High-level protein expression in scalable CHO transient transfection.", BIOTECHNOLOGY AND BIOENGINEERING, 2009, pages 542 - 551, XP055679536
JUSTIN BRYAN GOH ET AL.: "Impact of host cell line choice on glycan profile", CRITICAL REVIEWS IN BIOTECHNOLOGY, 2018, pages 851 - 867, XP055617047
KAMILLA SWIECH ET AL.: "Enhanced production of recombinant rabies virus glycoprotein (rRVGP) by Drosophila melanogaster S2 cells through control of culture conditions", CYTOTECHNOLOGY, 2008, pages 67 - 72, XP019602507
KYUNG HWA CHANG ET AL.: "Dimethylsulfoxide and sodium butyrate enhance the production of recombinant cyclooxygenase 2 in stably transformed Drosophila melanogaster S2 cells", BIOTECHNOLOGY LETTERS, 2002, pages 1353 - 1359, XP055679504
KYUNG HWA CHANG ET AL.: "Improved production of recombinant tumstatin in stably transformed Trichoplusia ni BTI Tn 5B1-4 cells", PROTEIN EXPRESSION AND PURIFICATION, 2004, pages 69 - 75, XP004497024
MELISSA HAMM ET AL.: "Characterization of N-Linked Glycosylation in a Monoclonal Antibody Produced in NS0 Cells Using Capillary Electrophoresis with Laser-Induced Fluorescence Detection", PHARMACEUTICALS, 2013, pages 393 - 406, XP009186031
MICHAEL F. WAHL ET AL.: "Effects of dimethyl sulfoxide on heavy chain monoclonal antibody production from plant cell culture", BIOTECHNOLOGY LETTERS, 1995, pages 463 - 468, XP055679501
P. HOSSLER ET AL: "Optimal and consistent protein glycosylation in mammalian cell culture", GLYCOBIOLOGY, vol. 19, no. 9, 3 June 2009 (2009-06-03), pages 936 - 949, XP055112498, ISSN: 0959-6658, DOI: 10.1093/glycob/cwp079 *
RICARDO GOUVEIA ET AL.: "Production and N-glycosylation of recombinant human cell adhesion molecule L1 from insect cells using the stable expression system. Effect of dimethyl sulfoxide", JOURNAL OF BIOTECHNOLOGY, 2010, pages 130 - 138, XP0026824485
SARAH F. SCHILLIE ET AL.: "Immune Response of Hepatitis B Vaccine Among Persons With Diabetes: A systematic review of the literature", DIABETES CARE, 2012, pages 2690 - 2697, XP055679619
SHUKLA A. K. ET AL.: "Dimethylsulphoxide as a tool to increase functional expression of heterologously produced GPCRs in mammalian cells", FEBS LETTERS, vol. 580, 2006, pages 4261 - 4265, XP005553322
THOMAS BERG ET AL.: "Purification and characterization of recombinant human lysosomal alpha-mannosidase", MOLECULAR GENETICS AND METABOLISM, vol. 73, 2001, pages 18 - 29, XP002182076
WENYING WANG ET AL.: "Gene transcription acceleration: main cause of hepatitis B surface antigen production improvement by dimethyl sulfoxide in the culture of Chinese hamster ovary cells", BIOTECHNOLOGY AND BIOENGINEERING, 2006, pages 526 - 535, XP055679518
XIA-JUAN XIA ET AL.: "Effects of inducers of differentiation on protein kinase C and CMP-N-acetylneuraminic acid:lactosylceramide sialyltransferase activities of HL-60 leukemia cells.", JOURNAL OF LIPID RESEARCH, 1989, pages 181 - 188, XP000941049
XIAO-LING LI ET AL.: "The preparation of HL-60 cells vaccine expressing BCG heat shock protein 70 and detection of its immunogenicity in vitro", HUMAN VACCINES & IMMUNOTHERAPEUTICS, 2012, pages 1376 - 1381, XP055679591

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11407813B2 (en) 2019-12-06 2022-08-09 Regeneron Pharmaceuticals, Inc. Anti-VEGF protein compositions and methods for producing the same
US11459373B2 (en) 2019-12-06 2022-10-04 Regeneron Pharmaceuticals, Inc. Anti-VEGF protein compositions and methods for producing the same
US11459374B2 (en) 2019-12-06 2022-10-04 Regeneron Pharmaceuticals, Inc. Anti-VEGF protein compositions and methods for producing the same
US11732025B2 (en) 2019-12-06 2023-08-22 Regeneron Pharmaceuticals, Inc. Anti-VEGF protein compositions and methods for producing the same

Also Published As

Publication number Publication date
US20140273057A1 (en) 2014-09-18
US8956830B2 (en) 2015-02-17
EP2968549A4 (en) 2016-08-17
US9487810B2 (en) 2016-11-08
US9926583B2 (en) 2018-03-27
US20170211114A1 (en) 2017-07-27
EP2968549B2 (en) 2022-11-23
US20150240281A1 (en) 2015-08-27
EP2968549A1 (en) 2016-01-20
ES2732679T3 (en) 2019-11-25
CN105073134A (en) 2015-11-18
WO2014150655A1 (en) 2014-09-25

Similar Documents

Publication Publication Date Title
US9926583B2 (en) Methods of cell culture
US10144944B2 (en) Methods of cell culture
US20170233782A1 (en) Methods of cell culture
US20240167069A1 (en) Sialylated Glycoproteins
US20140271622A1 (en) Methods of cell culture
US20220056109A1 (en) Glycoproteins with anti-inflammatory properties
US9914951B2 (en) Cell culture process

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20150730

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

A4 Supplementary search report drawn up and despatched

Effective date: 20160719

RIC1 Information provided on ipc code assigned before grant

Ipc: C12P 21/00 20060101ALI20160713BHEP

Ipc: A61K 39/395 20060101AFI20160713BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20170706

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602014046394

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: A61K0039395000

Ipc: C12P0021020000

RIC1 Information provided on ipc code assigned before grant

Ipc: C12P 21/02 20060101AFI20180219BHEP

Ipc: C12N 15/63 20060101ALI20180219BHEP

Ipc: G01N 33/68 20060101ALI20180219BHEP

Ipc: C07K 16/00 20060101ALI20180219BHEP

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MOMENTA PHARMACEUTICALS, INC.

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20180515

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTC Intention to grant announced (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20181025

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

Ref country code: AT

Ref legal event code: REF

Ref document number: 1130192

Country of ref document: AT

Kind code of ref document: T

Effective date: 20190515

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602014046394

Country of ref document: DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190908

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190808

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2732679

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20191125

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190808

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190809

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1130192

Country of ref document: AT

Kind code of ref document: T

Effective date: 20190508

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

REG Reference to a national code

Ref country code: DE

Ref legal event code: R026

Ref document number: 602014046394

Country of ref document: DE

PLBI Opposition filed

Free format text: ORIGINAL CODE: 0009260

PLAX Notice of opposition and request to file observation + time limit sent

Free format text: ORIGINAL CODE: EPIDOSNOBS2

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

26 Opposition filed

Opponent name: BAYER INTELLECTUAL PROPERTY GMBH

Effective date: 20200131

Opponent name: REGENERON PHARMACEUTICALS, INC.

Effective date: 20200131

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20200325

Year of fee payment: 7

PLBB Reply of patent proprietor to notice(s) of opposition received

Free format text: ORIGINAL CODE: EPIDOSNOBS3

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20200331

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200312

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200312

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200331

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210331

APBM Appeal reference recorded

Free format text: ORIGINAL CODE: EPIDOSNREFNO

APBP Date of receipt of notice of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA2O

APAH Appeal reference modified

Free format text: ORIGINAL CODE: EPIDOSCREFNO

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

APBU Appeal procedure closed

Free format text: ORIGINAL CODE: EPIDOSNNOA9O

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20220406

Year of fee payment: 9

PUAH Patent maintained in amended form

Free format text: ORIGINAL CODE: 0009272

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: PATENT MAINTAINED AS AMENDED

27A Patent maintained in amended form

Effective date: 20221123

AK Designated contracting states

Kind code of ref document: B2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: DE

Ref legal event code: R102

Ref document number: 602014046394

Country of ref document: DE

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602014046394

Country of ref document: DE

Representative=s name: MATHYS & SQUIRE EUROPE PATENTANWAELTE PARTNERS, DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20190508

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20230215

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20221123

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20190508

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IS

Payment date: 20240212

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20240130

Year of fee payment: 11

Ref country code: GB

Payment date: 20240201

Year of fee payment: 11